1
|
Fernandez-Flores A. Conceptual Contextualization of Neutrophilic Dermatoses. Am J Dermatopathol 2025; 47:337-354. [PMID: 39761647 DOI: 10.1097/dad.0000000000002836] [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: 04/19/2025]
Abstract
ABSTRACT Neutrophilic dermatoses are defined as inflammatory skin diseases characterized by sterile infiltration of polymorphonuclear neutrophils into various cutaneous layers. Although, in many cases, neutrophilic dermatoses represent the cutaneous counterpart of autoinflammatory diseases, this is not always the case, and there are other causes associated with this group of diseases, such as the administration of certain drugs or an underlying tumor. However, understanding the autoinflammatory context in which most of these entities develop, as well as their close relationship with autoimmunity, is key to comprehending their pathogenesis. In addition, understanding the mechanisms by which neutrophils migrate to the dermis and become activated is fundamental for interpreting the morphological findings of these biopsies. Finally, the description of a new group of neutrophilic dermatoses in recent years, in relation to keratinization disorders, has been crucial for understanding the best therapeutic approach for these difficult-to-manage entities.
Collapse
Affiliation(s)
- Angel Fernandez-Flores
- Dermatopathologist, Department of Cellular Pathology, Hospital Universitario El Bierzo, Ponferrada, Spain ; and
- Department of Cellular Pathology, Hospital de la Reina, Ponferrada, Spain
| |
Collapse
|
2
|
Labrecque MM, Allard MÈ, Murru A, Paré G, Acker JP, Lesage S, Girard M, Fernandes MJ. Prolonging neutrophil room-temperature storage with clinically approved solutions: implications for granulocyte transfusion. J Leukoc Biol 2025; 117:qiae258. [PMID: 39716952 DOI: 10.1093/jleuko/qiae258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/20/2024] [Indexed: 12/25/2024] Open
Abstract
Granulocyte concentrates (GCs) are leukocyte preparations enriched in neutrophils that can potentially save neutropenic patients from life-threatening, antimicrobial-resistant infections. The main challenge of GC transfusions is preserving the viability and antimicrobial activity of neutrophils beyond 24 h to reduce the logistical burden on collection centers and increase the availability of this cell therapy. Thus, the aim of this study was to explore extending the ex vivo viability and antimicrobial activity of GC neutrophils up to 72 h with a unique combination of the clinically approved additives Plasma-Lyte (PL), SAGM, AS-3, and Alburex. Neutrophils isolated from healthy donors were resuspended in autologous plasma at the same concentration as in GCs, diluted with various combinations of PL, SAGM, AS-3, and/or Alburex with or without the addition of buffers, and stored at room temperature for up to 72 h. During storage, neutrophil viability, phagocytosis, and intracellular reactive oxygen species production were measured by flow cytometry. Extracellular reactive oxygen species production was measured by spectrophotometry and chemotaxis by the number of calcein-stained neutrophils that migrated toward the chemotactic peptide, N-formyl-Met-Leu-Phe (fMLF). The same assays were performed on pooled, residual leukocyte units generated by the Reveos system, after storage in the additive combination that most effectively preserved the viability and function of isolated neutrophils. The additive combination that best performed in the majority of the assays contained PL, buffers, and AS-3. Neutrophil viability was preserved for a maximum of 48 h and phagocytosis of opsonized bacteria and reactive oxygen species production up to 72 h of storage at room temperature. In contrast, fMLF-induced chemotaxis decreased by 20% after 24-h storage while extracellular reactive oxygen species production increased significantly within the same time period. Supplementing GCs prepared from pooled, residual leukocyte units with this storage solution after the standard 16- to 24-h processing period as per the blood center guidelines, did not significantly improve the preservation of neutrophil viability and function. Our findings provide proof of concept that mixtures of clinically approved additives can be tailored to significantly prolong the viability and function of freshly isolated neutrophils during room-temperature storage. The unique additive composition of this storage solution that we developed for freshly isolated neutrophils requires further optimization for use with pooled, residual leukocyte units as well as the timepoint at which the solution is added during processing to prolong the viability and functions of neutrophils in this blood product.
Collapse
Affiliation(s)
- Marie-Michèle Labrecque
- Infectious and Immune Diseases Division, CHU de Québec Research Center, 2705 Blvd. Laurier, Québec, QC, G1V 4G2, Canada
- Faculty of Medicine, Department of Microbiology, Infectious Diseases and Immunology, Laval University, 1050 Av. de la Médecine, Québec, QC, G1V 0A6, Canada
- Medical Affairs and Innovation, Hema-Québec, 1070 Ave des Sciences-de-la-Vie, Québec, QC, G1V 5C3, Canada
| | - Marie-Ève Allard
- Medical Affairs and Innovation, Hema-Québec, 1070 Ave des Sciences-de-la-Vie, Québec, QC, G1V 5C3, Canada
| | - Andréa Murru
- Infectious and Immune Diseases Division, CHU de Québec Research Center, 2705 Blvd. Laurier, Québec, QC, G1V 4G2, Canada
- Faculty of Medicine, Department of Microbiology, Infectious Diseases and Immunology, Laval University, 1050 Av. de la Médecine, Québec, QC, G1V 0A6, Canada
- Medical Affairs and Innovation, Hema-Québec, 1070 Ave des Sciences-de-la-Vie, Québec, QC, G1V 5C3, Canada
| | - Guillaume Paré
- Infectious and Immune Diseases Division, CHU de Québec Research Center, 2705 Blvd. Laurier, Québec, QC, G1V 4G2, Canada
- Faculty of Medicine, Department of Microbiology, Infectious Diseases and Immunology, Laval University, 1050 Av. de la Médecine, Québec, QC, G1V 0A6, Canada
| | - Jason P Acker
- Innovation and Portfolio Management, Canadian Blood Services, 331 Canadian Blood Services, 8249 114 St NW, Edmonton, AB, T6G 2R8, Canada
- Department of Laboratory Medicine and Pathology, University of Alberta, 2-51B South Academic Building, 11328 - 89 Ave NW, Edmonton, AB, T6G 2J7, Canada
| | - Sylvie Lesage
- Immunology-Oncology Division, Maisonneuve-Rosemont Hospital Research Center, 5415 Bd de l'Assomption, Montréal, QC, H1T 2M4, Canada
- Département de microbiologie, infectiologie et immunologie, Université de Montréal, C.P. 6128, succ. Centre-ville, Montréal, QC, H3C 3J7, Canada
| | - Mélissa Girard
- Medical Affairs and Innovation, Hema-Québec, 1070 Ave des Sciences-de-la-Vie, Québec, QC, G1V 5C3, Canada
| | - Maria J Fernandes
- Infectious and Immune Diseases Division, CHU de Québec Research Center, 2705 Blvd. Laurier, Québec, QC, G1V 4G2, Canada
- Faculty of Medicine, Department of Microbiology, Infectious Diseases and Immunology, Laval University, 1050 Av. de la Médecine, Québec, QC, G1V 0A6, Canada
| |
Collapse
|
3
|
Rodriguez-Moncayo R, Pons S, Tavares LP, Jeon H, Preuss JA, Bahnemann J, Han J, Levy BD, Voldman J. Inertial Microfluidics Enables Functional Analysis of Neutrophils Isolated from Ultralow Blood Volume Samples. Anal Chem 2025; 97:8419-8428. [PMID: 40145542 DOI: 10.1021/acs.analchem.5c00102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
Monitoring immune cell function is increasingly being recognized as a more relevant biomarker than traditional white blood cell counts, yet the need for repeated relatively large blood volumes still continues to pose a significant challenge. To overcome this, we developed a sample-sparing platform using inertial microfluidics that can process as little as 10 μL of blood to isolate leukocytes for downstream functional analysis. Our platform isolates leukocytes with ∼80% purity, >90% in-device recovery, and >95% viability. Neutrophils were our primary focus due to their sensitivity to external stimuli and their critical role in immune responses. Neutrophils isolated through our new method did not show inadvertent activation, as evidenced by unchanged expression of activation markers CD62L and CD11b, with phenotypes comparable to control cells in whole blood. We conducted a range of functional assays, including phagocytosis, ROS production, and NETosis with all tests confirming that neutrophils maintained their functionality after isolation. These assays were performed using standard laboratory workflows, demonstrating the platform's compatibility with techniques such as flow cytometry and cell culture assays. Furthermore, we showed the versatility of our platform by successfully isolating leukocytes from challenging samples, including mouse blood from the vena cava or tail vein, as well as human capillary blood obtained by fingerstick. This adaptability highlights the potential of this platform for clinical and research applications, particularly in frequent immune monitoring or cases where sample volume is limited.
Collapse
Affiliation(s)
- Roberto Rodriguez-Moncayo
- Research Laboratory of Electronics, Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts 02139, United States
| | - Stephanie Pons
- Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Luciana P Tavares
- Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Hyungkook Jeon
- Research Laboratory of Electronics, Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts 02139, United States
- Department of Manufacturing Systems and Design Engineering (MSDE), Seoul National University of Science and Technology (SEOULTECH), 232 Gongneung-ro, Nowon-gu, Seoul 01811, Republic of Korea
| | | | - Janina Bahnemann
- Institute of Physics & Center for Advanced Analytics and Predictive Sciences, University of Augsburg, Augsburg 86159, Germany
| | - Jongyoon Han
- Research Laboratory of Electronics, Department of Electrical Engineering and Computer Science & Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts 02139, United States
| | - Bruce D Levy
- Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Joel Voldman
- Research Laboratory of Electronics, Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts 02139, United States
| |
Collapse
|
4
|
Lee S, Richman H, Yu Y. Nanocluster-mediated signaling crosstalk between FcγR and TLR4 in macrophage inflammatory responses. Sci Rep 2025; 15:12856. [PMID: 40229305 PMCID: PMC11997114 DOI: 10.1038/s41598-025-96679-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 03/31/2025] [Indexed: 04/16/2025] Open
Abstract
Receptor crosstalk, the interaction between different receptors to modulate signaling, is crucial for fine-tuning the inflammatory responses of innate immune cells. Although the synergistic crosstalk between Toll-like receptor (TLR)4 and Fc gamma receptor (FcγR) is well documented, the detailed mechanism underlying this synergy remains unclear. In this study, we addressed this knowledge gap by imaging the molecular organization of TLR4 and FcγR on the macrophage cell surface and correlating it with their synergistic co-activation using ligands functionalized on lipid bilayers. We confirmed that co-activation of TLR4 and FcγR enhances whole-cell pro-inflammatory responses and tyrosine phosphorylation at the receptor level. Super-resolution microscopy revealed that TLR4 and FcγR each form discrete nanoclusters after ligand stimulation, and their synergistic co-activation increases both the size and spatial overlap of these nanoclusters. Contrary to previous assumptions that TLR4 and FcγR form heterodimers during their crosstalk, our results emphasize the critical role of nanoscale spatial organization between distinct receptor clusters in modulating innate immune responses. Additionally, these findings align with similar receptor interaction mechanisms that we previously reported in other receptor pairs, such as Dectin-1/TLR2 and FcγR/TLR2, suggesting that nanocluster interactions may represent a predominant mechanism governing crosstalk between TLRs and ITAM-containing receptors.
Collapse
Affiliation(s)
- Seonik Lee
- Department of Chemistry, Indiana University, Bloomington, IN, USA
| | - Hunter Richman
- Department of Chemistry, Indiana University, Bloomington, IN, USA
| | - Yan Yu
- Department of Chemistry, Indiana University, Bloomington, IN, USA.
| |
Collapse
|
5
|
Bagatella S, Monney C, Gross N, Bernier Gosselin V, Schüpbach-Regula G, Hemphill A, Oevermann A. Intravacuolar persistence in neutrophils facilitates Listeria monocytogenes spread to co-cultured cells. mBio 2025; 16:e0270024. [PMID: 40067021 PMCID: PMC11980584 DOI: 10.1128/mbio.02700-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 02/10/2025] [Indexed: 04/10/2025] Open
Abstract
The bacterium Listeria monocytogenes (Lm) causes listeriosis in humans and ruminants. Acute lesions are predominantly infiltrated by polymorphonuclear neutrophils (PMNs), considered to be the efficient bactericidal arm of innate immunity. However, recent evidence suggests that PMNs cannot achieve antilisterial sterilizing immunity and that Lm may persist within PMNs. Despite this, interactions between PMNs and Lm remain poorly understood. In this study, we characterized the listericidal activity and interaction dynamics of bovine PMNs with Lm ex vivo. Phagocytosed Lm failed to escape into the PMN cytosol and was primarily targeted by phagolysosomal mechanisms. However, PMNs enabled prolonged intravacuolar survival of a resilient Lm subpopulation, largely as viable but non-culturable (VBNC) bacteria. This resilient Lm population could spread from PMNs to a cell line, resuscitate, and complete its canonical life cycle, thereby perpetuating the infection. Therefore, we identify PMNs as a mobile niche for Lm survival and provide evidence that PMNs harbor VBNC bacteria, potentially facilitating Lm dissemination within the host. IMPORTANCE Listeria monocytogenes (Lm) is a significant foodborne pathogen responsible for high hospitalization rates in humans, especially vulnerable groups such as the elderly, pregnant women, and immunocompromised individuals. In animals like ruminants, Lm infection leads to severe disease manifestations, notably brainstem encephalitis. This study uncovers a novel mechanism by which bovine neutrophils (PMNs) harbor Lm in a viable but non-culturable (VBNC) state, enabling the bacteria to hide in the host. PMNs, traditionally viewed as bacteria killers, may serve as Trojan horses, allowing Lm to persist and spread within the host. This discovery has broad implications for understanding Lm's persistence, its role in recurrent infections, and the development of new therapeutic strategies targeting VBNC forms of Lm to improve treatment outcomes and disease control.
Collapse
Affiliation(s)
- Stefano Bagatella
- Division of Neurological Sciences, NeuroCenter, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Camille Monney
- Division of Neurological Sciences, NeuroCenter, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Natascha Gross
- Division of Neurological Sciences, NeuroCenter, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | | | | | - Andrew Hemphill
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Anna Oevermann
- Division of Neurological Sciences, NeuroCenter, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
6
|
Santamarina-Fernández R, Fuentes-Valverde V, Silva-Rodríguez A, García P, Moscoso M, Bou G. Pseudomonas aeruginosa Vaccine Development: Lessons, Challenges, and Future Innovations. Int J Mol Sci 2025; 26:2012. [PMID: 40076637 PMCID: PMC11900337 DOI: 10.3390/ijms26052012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/18/2025] [Accepted: 02/23/2025] [Indexed: 03/14/2025] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen with a multidrug-resistant profile that has become a critical threat to global public health. It is one of the main causes of severe nosocomial infections, including ventilator-associated pneumonia, chronic infections in patients with cystic fibrosis, and bloodstream infections in immunosuppressed individuals. Development of vaccines against P. aeruginosa is a major challenge owing to the high capacity of this bacterium to form biofilms, its wide arsenal of virulence factors (including secretion systems, lipopolysaccharides, and outer membrane proteins), and its ability to evade the host immune system. This review provides a comprehensive historical overview of vaccine development efforts targeting this pathogen, ranging from early attempts in the 1970s to recent advancements, including vaccines based on novel proteins and emerging technologies such as nanoparticles and synthetic conjugates. Despite numerous promising preclinical developments, very few candidates have progressed to clinical trials, and none have achieved final approval. This panorama highlights the significant scientific efforts undertaken and the inherent complexity of successfully developing an effective vaccine against P. aeruginosa.
Collapse
Affiliation(s)
- Rebeca Santamarina-Fernández
- Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain; (R.S.-F.); (V.F.-V.); (A.S.-R.); (P.G.); (G.B.)
| | - Víctor Fuentes-Valverde
- Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain; (R.S.-F.); (V.F.-V.); (A.S.-R.); (P.G.); (G.B.)
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Área de Medicamentos Biológicos, Agencia Española de Medicamentos y Productos Sanitarios (AEMPS), 28022 Madrid, Spain
| | - Alis Silva-Rodríguez
- Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain; (R.S.-F.); (V.F.-V.); (A.S.-R.); (P.G.); (G.B.)
| | - Patricia García
- Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain; (R.S.-F.); (V.F.-V.); (A.S.-R.); (P.G.); (G.B.)
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Miriam Moscoso
- Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain; (R.S.-F.); (V.F.-V.); (A.S.-R.); (P.G.); (G.B.)
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Germán Bou
- Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain; (R.S.-F.); (V.F.-V.); (A.S.-R.); (P.G.); (G.B.)
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Universidad de A Coruña, 15006 A Coruña, Spain
| |
Collapse
|
7
|
Rodrigues RRL, de Sousa JMS, Dos Santos ALS, de Souza VMR, Machado YAA, de Lima Nunes TA, da Silva MV, de Araújo-Nobre AR, de Araújo RSA, Mendonça Junior FJB, Veras LMDC, Rodrigues KADF. Evaluation of SB-83, a 2-amino-thiophene derivative, against Leishmania species that cause visceral leishmaniasis. Int Immunopharmacol 2025; 148:114106. [PMID: 39862638 DOI: 10.1016/j.intimp.2025.114106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/06/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
Visceral leishmaniasis is a systemic disease that affects various internal organs and represents the most severe and fatal form of leishmaniasis. Conventional treatment presents significant challenges, such as prolonged management in hospital settings, high toxicity, and an increasing growing number of cases of resistance. In previous studies, our research group demonstrated the effective and selective activity of the 2-amino-thiophene derivative SB-83 in preclinical models of cutaneous leishmaniasis. Given the urgent need for new therapeutic alternatives for visceral leishmaniasis, and considering our previous promising results for SB-83, this study investigated the antileishmanial activity of the compound on the etiological agents of visceral leishmaniasis. SB-83 demonstrated efficacy in inhibiting the growth of promastigote forms of Leishmania (Leishmania) infantum (IC50 = 7.46 µM) and Leishmania (Leishmania) donovani (IC50 = 9.84 µM). In the cytotoxicity evaluation, in RAW 264.7 macrophages, the compound revealed a CC50 = 52.27 µM, being more toxic to the parasite, and respective selectivity indices (SI) of 7 and 5.31 against the previously mentioned species. Atomic force microscopy analysis showed that the compound causes alterations in surface roughness, formation of englobulations, and accumulation of lipids, all of which are indicative of cell death by apoptosis. This was confirmed by flow cytometry, which showed an increase in the number of cells labeled with Annexin V-FITC+/IP-, indicating apoptosis. SB-83 showed even greater efficacy against intramacrophagic amastigote forms (EC50 = 2.91 µM), which was associated with structural changes, such as increased lysosomal volume, and cellular mechanisms, including elevated levels of cytokines TNF-α and IL-12, reactive oxygen and nitrogen species, and reduced levels of cytokines IL-10 and IL-6, as well as decreased arginase activity. The results allow us to conclude that the 2-amino-thiophene derivative SB-83 is a promising compound for development of new treatments against visceral leishmaniasis.
Collapse
Affiliation(s)
- Raiza Raianne Luz Rodrigues
- Infectious Diseases Laboratory, Campus Ministro Reis Velloso, Federal University of Parnaíba Delta, 64202-020 Parnaíba, PI, Brazil
| | - Julyanne Maria Saraiva de Sousa
- Infectious Diseases Laboratory, Campus Ministro Reis Velloso, Federal University of Parnaíba Delta, 64202-020 Parnaíba, PI, Brazil
| | - Airton Lucas Sousa Dos Santos
- Infectious Diseases Laboratory, Campus Ministro Reis Velloso, Federal University of Parnaíba Delta, 64202-020 Parnaíba, PI, Brazil
| | - Vanessa Maria Rodrigues de Souza
- Infectious Diseases Laboratory, Campus Ministro Reis Velloso, Federal University of Parnaíba Delta, 64202-020 Parnaíba, PI, Brazil
| | - Yasmim Alves Aires Machado
- Infectious Diseases Laboratory, Campus Ministro Reis Velloso, Federal University of Parnaíba Delta, 64202-020 Parnaíba, PI, Brazil
| | - Thaís Amanda de Lima Nunes
- Laboratory of Immunology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, 38025-180 Uberaba, MG, Brazil
| | - Marcos Vinícius da Silva
- Laboratory of Immunology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, 38025-180 Uberaba, MG, Brazil
| | - Alyne Rodrigues de Araújo-Nobre
- Biodiversity and Biotechnology Research Center, Campus Ministro Reis Velloso, Federal University of Parnaíba Delta, 64202-020 Parnaíba, PI, Brazil
| | - Rodrigo Santos Aquino de Araújo
- Laboratory of Synthesis and Drug Delivery, Department of Biological Sciences, State University of Paraíba, 58071-160 João Pessoa, PB, Brazil
| | | | - Leiz Maria da Costa Veras
- Biodiversity and Biotechnology Research Center, Campus Ministro Reis Velloso, Federal University of Parnaíba Delta, 64202-020 Parnaíba, PI, Brazil
| | | |
Collapse
|
8
|
Wilton ZER, Jamus AN, Core SB, Frietze KM. Pathogenic and Protective Roles of Neutrophils in Chlamydia trachomatis Infection. Pathogens 2025; 14:112. [PMID: 40005489 PMCID: PMC11858174 DOI: 10.3390/pathogens14020112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/14/2025] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
Chlamydia trachomatis (Ct) is an obligate intracellular pathogen that causes the most commonly diagnosed bacterial sexually transmitted infection (STI) and is a leading cause of preventable blindness globally. Ct infections can generate a strong pro-inflammatory immune response, leading to immune-mediated pathology in infected tissues. Neutrophils play an important role in mediating both pathology and protection during infection. Excessive neutrophil activation, migration, and survival are associated with host tissue damage during Chlamydia infections. In contrast, neutrophils also perform phagocytic killing of Chlamydia in the presence of IFN-γ and anti-Chlamydia antibodies. Neutrophil extracellular traps (NETs) and many neutrophil degranulation products have also demonstrated strong anti-Chlamydia functions. To counteract this neutrophil-mediated protection, Chlamydia has developed several evasion strategies. Various Chlamydia proteins can limit potentially protective neutrophil responses by directly targeting receptors present on the surface of neutrophils or neutrophil degranulation products. In this review, we provide a survey of current knowledge regarding the role of neutrophils in pathogenesis and protection, including the ways that Chlamydia circumvents neutrophil functions, and we propose critical areas for future research.
Collapse
Affiliation(s)
| | | | | | - Kathryn M. Frietze
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences, Albuquerque, NM 87131, USA
| |
Collapse
|
9
|
Wang Z, Saxena A, Yan W, Uriarte SM, Siqueira R, Li X. The impact of aging on neutrophil functions and the contribution to periodontitis. Int J Oral Sci 2025; 17:10. [PMID: 39819982 PMCID: PMC11739572 DOI: 10.1038/s41368-024-00332-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/21/2024] [Accepted: 10/21/2024] [Indexed: 01/19/2025] Open
Abstract
The increasing aging population and aging-associated diseases have become a global issue for decades. People over 65 show an increased prevalence and greater severity of periodontitis, which poses threats to overall health. Studies have demonstrated a significant association between aging and the dysfunction of neutrophils, critical cells in the early stages of periodontitis, and their crosstalk with macrophages and T and B lymphocytes to establish the periodontal lesion. Neutrophils differentiate and mature in the bone marrow before entering the circulation; during an infection, they are recruited to infected tissues guided by the signal from chemokines and cytokines to eliminate invading pathogens. Neutrophils are crucial in maintaining a balanced response between host and microbes to prevent periodontal diseases in periodontal tissues. The impacts of aging on neutrophils' chemotaxis, anti-microbial function, cell activation, and lifespan result in impaired neutrophil functions and excessive neutrophil activation, which could influence periodontitis course. We summarize the roles of neutrophils in periodontal diseases and the aging-related impacts on neutrophil functional responses. We also explore the underlying mechanisms that can contribute to periodontitis manifestation in aging. This review could help us better understand the pathogenesis of periodontitis, which could offer novel therapeutic targets for periodontitis.
Collapse
Affiliation(s)
- Zi Wang
- Department of Plastic Surgery, Maxillofacial & Oral Health, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Anish Saxena
- Molecular Pathobiology Department, New York University College of Dentistry, New York, NY, USA
| | - Wenbo Yan
- Department of Plastic Surgery, Maxillofacial & Oral Health, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Silvia M Uriarte
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, KY, USA
| | - Rafael Siqueira
- Department of Periodontics, Virginia Commonwealth University School of Dentistry, Richmond, VA, USA
| | - Xin Li
- Department of Plastic Surgery, Maxillofacial & Oral Health, University of Virginia School of Medicine, Charlottesville, VA, USA.
- Molecular Pathobiology Department, New York University College of Dentistry, New York, NY, USA.
- Comprehensive Cancer Center, University of Virginia, Charlottesville, USA.
| |
Collapse
|
10
|
Hitzler SUJ, Fernández-Fernández C, Montaño DE, Dietschmann A, Gresnigt MS. Microbial adaptive pathogenicity strategies to the host inflammatory environment. FEMS Microbiol Rev 2025; 49:fuae032. [PMID: 39732621 PMCID: PMC11737513 DOI: 10.1093/femsre/fuae032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/19/2024] [Accepted: 12/27/2024] [Indexed: 12/30/2024] Open
Abstract
Pathogenic microorganisms can infect a variety of niches in the human body. During infection, these microbes can only persist if they adapt adequately to the dynamic host environment and the stresses imposed by the immune system. While viruses entirely rely on host cells to replicate, bacteria and fungi use their pathogenicity mechanisms for the acquisition of essential nutrients that lie under host restriction. An inappropriate deployment of pathogenicity mechanisms will alert host defence mechanisms that aim to eradicate the pathogen. Thus, these adaptations require tight regulation to guarantee nutritional access without eliciting strong immune activation. To work efficiently, the immune system relies on a complex signalling network, involving a myriad of immune mediators, some of which are quite directly associated with imminent danger for the pathogen. To manipulate the host immune system, viruses have evolved cytokine receptors and viral cytokines. However, among bacteria and fungi, selected pathogens have evolved the capacity to use these inflammatory response-specific signals to regulate their pathogenicity. In this review, we explore how bacterial and fungal pathogens can sense the immune system and use adaptive pathogenicity strategies to evade and escape host defence to ensure their persistence in the host.
Collapse
Affiliation(s)
- Sophia U J Hitzler
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute, 07745 Jena, Germany
| | - Candela Fernández-Fernández
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute, 07745 Jena, Germany
| | - Dolly E Montaño
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute, 07745 Jena, Germany
| | - Axel Dietschmann
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute, 07745 Jena, Germany
| | - Mark S Gresnigt
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute, 07745 Jena, Germany
| |
Collapse
|
11
|
Bolduan V, Palzer KA, Ries F, Busch N, Pautz A, Bros M. KSRP Deficiency Attenuates the Course of Pulmonary Aspergillosis and Is Associated with the Elevated Pathogen-Killing Activity of Innate Myeloid Immune Cells. Cells 2024; 13:2040. [PMID: 39768132 PMCID: PMC11674352 DOI: 10.3390/cells13242040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/12/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
The mRNA-binding protein KSRP (KH-type splicing regulatory protein) is known to modulate immune cell functions post-transcriptionally, e.g., by reducing the mRNA stability of cytokines. It is known that KSRP binds the AU-rich motifs (ARE) that are often located in the 3'-untranslated part of mRNA species, encoding dynamically regulated proteins as, for example, cytokines. Innate myeloid immune cells, such as polymorphonuclear neutrophils (PMNs) and macrophages (MACs), eliminate pathogens by multiple mechanisms, including phagocytosis and the secretion of chemo- and cytokines. Here, we investigated the role of KSRP in the phenotype and functions of both innate immune cell types in the mouse model of invasive pulmonary aspergillosis (IPA). Here, KSRP-/- mice showed lower levels of Aspergillus fumigatus conidia (AFC) and an increase in the frequencies of PMNs and MACs in the lungs. Our results showed that PMNs and MACs from KSRP-/- mice exhibited an enhanced phagocytic uptake of AFC, accompanied by increased ROS production in PMNs upon stimulation. A comparison of RNA sequencing data revealed that 64 genes related to inflammatory and immune responses were shared between PMNs and MACs. The majority of genes upregulated in PMNs were involved in metabolic processes, cell cycles, and DNA repair. Similarly, KSRP-deficient PMNs displayed reduced levels of apoptosis. In conclusion, our results indicate that KSRP serves as a critical negative regulator of PMN and MAC anti-pathogen activity.
Collapse
Affiliation(s)
- Vanessa Bolduan
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Kim-Alicia Palzer
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Frederic Ries
- Department of Hematology and Medical Oncology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Nora Busch
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Andrea Pautz
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Matthias Bros
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| |
Collapse
|
12
|
Herdendorf TJ, Mishra N, Fatehi S, Gido CD, Prakash O, Geisbrecht BV. New advances in understanding inhibition of myeloperoxidase and neutrophil serine proteases by two families of staphylococcal innate immune evasion proteins. Arch Biochem Biophys 2024; 761:110177. [PMID: 39393662 PMCID: PMC11560548 DOI: 10.1016/j.abb.2024.110177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/25/2024] [Accepted: 10/08/2024] [Indexed: 10/13/2024]
Abstract
Neutrophils are the most abundant leukocytes in humans and play an important early role in the innate immune response against microorganisms. Neutrophil phagosomes contain high concentrations of antibacterial enzymes, including myeloperoxidase (MPO) and the neutrophil serine proteases (NSPs). These antibacterial enzymes can also be released extracellularly upon degranulation or as a component of neutrophil extracellular traps (NETs). Due to host/pathogen coevolution, S. aureus expresses a diverse arsenal of innate immune evasion proteins that target many aspects of the neutrophil antibacterial response. In the last decade, two new classes of staphylococcal innate immune evasion proteins that act as potent, selective inhibitors of MPO and NSPs, respectively, have been discovered. The Staphylococcal Peroxidase INhibitor (SPIN) is a small ∼8.3 kDa α-helical bundle protein that blocks MPO activity by interfering with substrate and product exchange with the MPO active site. The Extracellular Adherence Protein (EAP) family consists of three unique proteins comprised of one or more copies of an ∼11 kDa β-grasp domain capable of high-affinity, selective, non-covalent inhibition of NSPs. This brief review article summarizes recent advances in understanding the structural and functional properties of SPIN and EAP family members and outlines some potential avenues for future investigation of these enzyme inhibitors.
Collapse
Affiliation(s)
- Timothy J Herdendorf
- Department of Biochemistry & Molecular Biophysics, Kansas State University, Manhattan, KS 66506, USA
| | - Nitin Mishra
- Department of Biochemistry & Molecular Biophysics, Kansas State University, Manhattan, KS 66506, USA
| | - Soheila Fatehi
- Department of Biochemistry & Molecular Biophysics, Kansas State University, Manhattan, KS 66506, USA
| | - Carson D Gido
- Department of Biochemistry & Molecular Biophysics, Kansas State University, Manhattan, KS 66506, USA
| | - Om Prakash
- Department of Biochemistry & Molecular Biophysics, Kansas State University, Manhattan, KS 66506, USA.
| | - Brian V Geisbrecht
- Department of Biochemistry & Molecular Biophysics, Kansas State University, Manhattan, KS 66506, USA.
| |
Collapse
|
13
|
Yoon G, Puentes R, Tran J, Multani A, Cobo ER. The role of cathelicidins in neutrophil biology. J Leukoc Biol 2024; 116:689-705. [PMID: 38758953 DOI: 10.1093/jleuko/qiae112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/14/2024] [Accepted: 04/25/2024] [Indexed: 05/19/2024] Open
Abstract
Despite their relatively short lifespan, neutrophils are tasked with counteracting pathogens through various functions, including phagocytosis, production of reactive oxygen species, neutrophil extracellular traps (NETs), and host defense peptides. Regarding the latter, small cationic cathelicidins present a conundrum in neutrophil function. Although primarily recognized as microbicides with an ability to provoke pores in microbial cell walls, the ability of cathelicidin to modulate key neutrophil functions is also of great importance, including the release of chemoattractants, cytokines, and reactive oxygen species, plus prolonging neutrophil lifespan. Cumulative evidence indicates a less recognized role of cathelicidin as an "immunomodulator"; however, this term is not always explicit, and its relevance in neutrophil responses during infection and inflammation is seldom discussed. This review compiles and discusses studies of how neutrophils use cathelicidin to respond to infections, while also acknowledging immunomodulatory aspects of cathelicidin through potential crosstalk between sources of the peptide.
Collapse
Affiliation(s)
- Grace Yoon
- Faculty of Veterinary Medicine, University of Calgary, HSC 1871, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada
| | - Rodrigo Puentes
- Faculty of Veterinary Medicine, University of Calgary, HSC 1871, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada
| | - Jacquelyn Tran
- Faculty of Veterinary Medicine, University of Calgary, HSC 1871, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada
| | - Anmol Multani
- Faculty of Veterinary Medicine, University of Calgary, HSC 1871, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada
| | - Eduardo R Cobo
- Faculty of Veterinary Medicine, University of Calgary, HSC 1871, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
14
|
Held J, Sivaraman K, Wrenger S, Si W, Welte T, Immenschuh S, Janciauskiene S. Ex vivo study on the human blood neutrophil circadian features and effects of alpha1-antitrypsin and lipopolysaccharide. Vascul Pharmacol 2024; 156:107396. [PMID: 38897556 DOI: 10.1016/j.vph.2024.107396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 06/10/2024] [Accepted: 06/16/2024] [Indexed: 06/21/2024]
Abstract
AIMS Neutrophils perform various functions in a circadian-dependent manner; therefore, we investigated here whether the effect of alpha1-antitrypsin (AAT), used as augmentation therapy, is dependent on the neutrophil circadian clock. AAT is a vital regulator of neutrophil functions, and its qualitative and/or quantitative defects have significant implications for the development of respiratory diseases. METHODS Whole blood from 12 healthy women age years, mean (SD) 29.92 (5.48) was collected twice daily, 8 h apart, and incubated for 30 min at 37 °C alone or with additions of 2 mg/ml AAT (Respreeza) and/or 5 μg/ml lipopolysaccharide (LPS) from Escherichia coli. Neutrophils were then isolated to examine gene expression, migration and phagocytosis. RESULTS The expression of CD14, CD16, CXCR2 and SELL (encoding CD62L) genes was significantly higher while CDKN1A lower in the afternoon than in the morning neutrophils from untreated blood. Neutrophils isolated in the afternoon had higher migratory and phagocytic activity. Morning neutrophils isolated from AAT-pretreated blood showed higher expression of CXCR2 and SELL than those from untreated morning blood. Pretreatment of blood with AAT enhanced migratory properties of morning but not afternoon neutrophils. Of all genes analysed, only CXCL8 expression was strongly upregulated in morning and afternoon neutrophils isolated from LPS-pretreated blood, whereas CXCR2 expression was downregulated in afternoon neutrophils. The addition of AAT did not reverse the effects of LPS. SIGNIFICANCE The circadian clock of myeloid cells may affect the effectiveness of various therapies, including AAT therapy used to treat patients with AAT deficiency, and needs further investigation.
Collapse
Affiliation(s)
- Julia Held
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany; Hannover Medical School, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| | - Kokilavani Sivaraman
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany; Hannover Medical School, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| | - Sabine Wrenger
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany; Hannover Medical School, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| | - Wenzhang Si
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany; Hannover Medical School, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| | - Tobias Welte
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany; Hannover Medical School, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| | - Stephan Immenschuh
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Sabina Janciauskiene
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany; Hannover Medical School, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany.
| |
Collapse
|
15
|
Liang Y, Wu G, Tan J, Xiao X, Yang L, Saw PE. Targeting NETosis: nature's alarm system in cancer progression. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:28. [PMID: 39143953 PMCID: PMC11322967 DOI: 10.20517/cdr.2024.24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/30/2024] [Accepted: 07/15/2024] [Indexed: 08/16/2024]
Abstract
Neutrophils are recognized active participants in inflammatory responses and are intricately linked to cancer progression. In response to inflammatory stimuli, neutrophils become activated, releasing neutrophils extracellular traps (NETs) for the capture and eradication of pathogens, a phenomenon termed NETosis. With a deeper understanding of NETs, there is growing evidence supporting their role in cancer progression and their involvement in conferring resistance to various cancer therapies, especially concerning tumor reactions to chemotherapy, radiation therapy (RT), and immunotherapy. This review summarizes the roles of NETs in the tumor microenvironment (TME) and their mechanisms of neutrophil involvement in the host defense. Additionally, it elucidates the mechanisms through which NETs promote tumor progression and their role in cancer treatment resistance, highlighting their potential as promising therapeutic targets in cancer treatment and their clinical applicability.
Collapse
Affiliation(s)
- Yixia Liang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, Guangdong, China
- Authors contributed equally
| | - Guo Wu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, Guangdong, China
- Authors contributed equally
| | - Jiabao Tan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, Guangdong, China
| | - Xiaoyun Xiao
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, Guangdong, China
| | - Linbin Yang
- Breast Tumor Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, Guangdong, China
| | - Phei Er Saw
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, Guangdong, China
| |
Collapse
|
16
|
Sun C, Wang S, Ma Z, Zhou J, Ding Z, Yuan G, Pan Y. Neutrophils in glioma microenvironment: from immune function to immunotherapy. Front Immunol 2024; 15:1393173. [PMID: 38779679 PMCID: PMC11109384 DOI: 10.3389/fimmu.2024.1393173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 04/25/2024] [Indexed: 05/25/2024] Open
Abstract
Glioma is a malignant tumor of the central nervous system (CNS). Currently, effective treatment options for gliomas are still lacking. Neutrophils, as an important member of the tumor microenvironment (TME), are widely distributed in circulation. Recently, the discovery of cranial-meningeal channels and intracranial lymphatic vessels has provided new insights into the origins of neutrophils in the CNS. Neutrophils in the brain may originate more from the skull and adjacent vertebral bone marrow. They cross the blood-brain barrier (BBB) under the action of chemokines and enter the brain parenchyma, subsequently migrating to the glioma TME and undergoing phenotypic changes upon contact with tumor cells. Under glycolytic metabolism model, neutrophils show complex and dual functions in different stages of cancer progression, including participation in the malignant progression, immune suppression, and anti-tumor effects of gliomas. Additionally, neutrophils in the TME interact with other immune cells, playing a crucial role in cancer immunotherapy. Targeting neutrophils may be a novel generation of immunotherapy and improve the efficacy of cancer treatments. This article reviews the molecular mechanisms of neutrophils infiltrating the central nervous system from the external environment, detailing the origin, functions, classifications, and targeted therapies of neutrophils in the context of glioma.
Collapse
Affiliation(s)
- Chao Sun
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Neurology of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Siwen Wang
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Zhen Ma
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Neurology of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Jinghuan Zhou
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Neurology of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Zilin Ding
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Neurology of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Guoqiang Yuan
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Neurology of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Yawen Pan
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Neurology of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
17
|
Yuan S, Hu Q. Convergence of nanomedicine and neutrophils for drug delivery. Bioact Mater 2024; 35:150-166. [PMID: 38318228 PMCID: PMC10839777 DOI: 10.1016/j.bioactmat.2024.01.022] [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: 10/31/2023] [Revised: 01/20/2024] [Accepted: 01/21/2024] [Indexed: 02/07/2024] Open
Abstract
Neutrophils have recently emerged as promising carriers for drug delivery due to their unique properties including rapid response toward inflammation, chemotaxis, and transmigration. When integrated with nanotechnology that has enormous advantages in improving treatment efficacy and reducing side effects, neutrophil-based nano-drug delivery systems have expanded the repertoire of nanoparticles employed in precise therapeutic interventions by either coating nanoparticles with their membranes, loading nanoparticles inside living cells, or engineering chimeric antigen receptor (CAR)-neutrophils. These neutrophil-inspired therapies have shown superior biocompatibility, targeting ability, and therapeutic robustness. In this review, we summarized the benefits of combining neutrophils and nanotechnologies, the design principles and underlying mechanisms, and various applications in disease treatments. The challenges and prospects for neutrophil-based drug delivery systems were also discussed.
Collapse
Affiliation(s)
- Sichen Yuan
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, United States
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, United States
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, United States
| | - Quanyin Hu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, United States
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, United States
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, United States
| |
Collapse
|
18
|
Mandelli AP, Magri G, Tortoli M, Torricelli S, Laera D, Bagnoli F, Finco O, Bensi G, Brazzoli M, Chiarot E. Vaccination with staphylococcal protein A protects mice against systemic complications of skin infection recurrences. Front Immunol 2024; 15:1355764. [PMID: 38529283 PMCID: PMC10961379 DOI: 10.3389/fimmu.2024.1355764] [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: 12/14/2023] [Accepted: 02/16/2024] [Indexed: 03/27/2024] Open
Abstract
Skin and soft tissue infections (SSTIs) are the most common diseases caused by Staphylococcus aureus (S. aureus), which can progress to threatening conditions due to recurrences and systemic complications. Staphylococcal protein A (SpA) is an immunomodulator antigen of S. aureus, which allows bacterial evasion from the immune system by interfering with different types of immune responses to pathogen antigens. Immunization with SpA could potentially unmask the pathogen to the immune system, leading to the production of antibodies that can protect from a second encounter with S. aureus, as it occurs in skin infection recurrences. Here, we describe a study in which mice are immunized with a mutated form of SpA mixed with the Adjuvant System 01 (SpAmut/AS01) before a primary S. aureus skin infection. Although mice are not protected from the infection under these conditions, they are able to mount a broader pathogen-specific functional immune response that results in protection against systemic dissemination of bacteria following an S. aureus second infection (recurrence). We show that this "hidden effect" of SpA can be partially explained by higher functionality of induced anti-SpA antibodies, which promotes better phagocytic activity. Moreover, a broader and stronger humoral response is elicited against several S. aureus antigens that during an infection are masked by SpA activity, which could prevent S. aureus spreading from the skin through the blood.
Collapse
Affiliation(s)
| | - Greta Magri
- Bacterial Vx Unit, GlaxoSmithKline, Siena, Italy
| | - Marco Tortoli
- Animal Resource Center, GlaxoSmithKline, Siena, Italy
| | | | | | - Fabio Bagnoli
- Infectious Disease Research Unit, GlaxoSmithKline, Upper Providence, PA, United States
| | - Oretta Finco
- Bacterial Vx Unit, GlaxoSmithKline, Siena, Italy
| | | | | | | |
Collapse
|
19
|
Awasthi D, Sarode A. Neutrophils at the Crossroads: Unraveling the Multifaceted Role in the Tumor Microenvironment. Int J Mol Sci 2024; 25:2929. [PMID: 38474175 DOI: 10.3390/ijms25052929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
Over the past decade, research has prominently established neutrophils as key contributors to the intricate landscape of tumor immune biology. As polymorphonuclear granulocytes within the innate immune system, neutrophils play a pivotal and abundant role, constituting approximately ∼70% of all peripheral leukocytes in humans and ∼10-20% in mice. This substantial presence positions them as the frontline defense against potential threats. Equipped with a diverse array of mechanisms, including reactive oxygen species (ROS) generation, degranulation, phagocytosis, and the formation of neutrophil extracellular traps (NETs), neutrophils undeniably serve as indispensable components of the innate immune system. While these innate functions enable neutrophils to interact with adaptive immune cells such as T, B, and NK cells, influencing their functions, they also engage in dynamic interactions with rapidly dividing tumor cells. Consequently, neutrophils are emerging as crucial regulators in both pro- and anti-tumor immunity. This comprehensive review delves into recent research to illuminate the multifaceted roles of neutrophils. It explores their diverse functions within the tumor microenvironment, shedding light on their heterogeneity and their impact on tumor recruitment, progression, and modulation. Additionally, the review underscores their potential anti-tumoral capabilities. Finally, it provides valuable insights into clinical therapies targeting neutrophils, presenting a promising approach to leveraging innate immunity for enhanced cancer treatment.
Collapse
Affiliation(s)
- Deepika Awasthi
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Aditya Sarode
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| |
Collapse
|
20
|
Mukhopadhyay A, Tsukasaki Y, Chan WC, Le JP, Kwok ML, Zhou J, Natarajan V, Mostafazadeh N, Maienschein-Cline M, Papautsky I, Tiruppathi C, Peng Z, Rehman J, Ganesh B, Komarova Y, Malik AB. trans-Endothelial neutrophil migration activates bactericidal function via Piezo1 mechanosensing. Immunity 2024; 57:52-67.e10. [PMID: 38091995 PMCID: PMC10872880 DOI: 10.1016/j.immuni.2023.11.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 08/02/2023] [Accepted: 11/10/2023] [Indexed: 12/21/2023]
Abstract
The regulation of polymorphonuclear leukocyte (PMN) function by mechanical forces encountered during their migration across restrictive endothelial cell junctions is not well understood. Using genetic, imaging, microfluidic, and in vivo approaches, we demonstrated that the mechanosensor Piezo1 in PMN plasmalemma induced spike-like Ca2+ signals during trans-endothelial migration. Mechanosensing increased the bactericidal function of PMN entering tissue. Mice in which Piezo1 in PMNs was genetically deleted were defective in clearing bacteria, and their lungs were predisposed to severe infection. Adoptive transfer of Piezo1-activated PMNs into the lungs of Pseudomonas aeruginosa-infected mice or exposing PMNs to defined mechanical forces in microfluidic systems improved bacterial clearance phenotype of PMNs. Piezo1 transduced the mechanical signals activated during transmigration to upregulate nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 4, crucial for the increased PMN bactericidal activity. Thus, Piezo1 mechanosensing of increased PMN tension, while traversing the narrow endothelial adherens junctions, is a central mechanism activating the host-defense function of transmigrating PMNs.
Collapse
Affiliation(s)
- Amitabha Mukhopadhyay
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Yoshikazu Tsukasaki
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Wan Ching Chan
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Jonathan P Le
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Man Long Kwok
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Jian Zhou
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois, Chicago, IL 60612, USA
| | - Viswanathan Natarajan
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA; Department of Medicine, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Nima Mostafazadeh
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois, Chicago, IL 60612, USA
| | - Mark Maienschein-Cline
- Research Informatics Core, Research Resources Center, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Ian Papautsky
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois, Chicago, IL 60612, USA
| | - Chinnaswamy Tiruppathi
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Zhangli Peng
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois, Chicago, IL 60612, USA
| | - Jalees Rehman
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Balaji Ganesh
- Flow Cytometry Core, Research Resources Center, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Yulia Komarova
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA.
| | - Asrar B Malik
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA.
| |
Collapse
|
21
|
Ouyang J, Hong Y, Wan Y, He X, Geng B, Yang X, Xiang J, Cai J, Zeng Z, Liu Z, Peng N, Jiang Y, Liu J. PVB exerts anti-inflammatory effects by inhibiting the activation of MAPK and NF-κB signaling pathways and ROS generation in neutrophils. Int Immunopharmacol 2024; 126:111271. [PMID: 38006749 DOI: 10.1016/j.intimp.2023.111271] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/13/2023] [Accepted: 11/17/2023] [Indexed: 11/27/2023]
Abstract
Pinaverium bromide (PVB) has been shown to protect mice against sepsis, which is predominantly attributed to PVB-mediated anti-inflammatory effects by inhibiting primed neutrophils to produce proinflammatory cytokines. However, the underlying mechanism(s) by which PVB affects neutrophils remains unknown. In this study, we report that treatment with PVB either before or after LPS stimulation attenuated IL-1β and TNF-α expression at both mRNA and protein levels in LPS-activated murine neutrophils. Further experiments revealed that PVB inhibited the phosphorylation of ERK, JNK, and IκBα in LPS-stimulated murine neutrophils. Moreover, PVB reduced reactive oxygen species (ROS) levels via regulating NADPH oxidase 2 (NOX2) activity, as represented by inhibiting p47phox translocation from the cytoplasm to the cellular membrane. Importantly, PVB significantly attenuated IL-1β, TNF-α, IL-6, CXCL1 production in both LPS-stimulated low density neutrophils (LDNs) and normal density neutrophils (NDNs) isolated from septic patients. Collectively, we demonstrated that PVB exerts anti-inflammatory effect by attenuating ROS generation and suppressing the activation of MAPK and NF-κB signaling pathways, suggesting that PVB may act as a potential therapeutic agent for sepsis by inhibiting neutrophil priming and activation.
Collapse
Affiliation(s)
- Jiafu Ouyang
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yinghao Hong
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yantong Wan
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiangyi He
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Bingxuan Geng
- School of the First Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Xinxing Yang
- School of the First Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Jing Xiang
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Junwei Cai
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhenhua Zeng
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhifeng Liu
- Department of Critical Care Medicine, General Hospital of Southern Theater Command, Guangzhou, Guangdong, China
| | - Na Peng
- Department of Emergency Medicine, General Hospital of Southern Theater Command, Guangzhou, Guangdong, China.
| | - Yong Jiang
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China.
| | - Jinghua Liu
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
22
|
Salken I, Provencio JJ, Coulibaly AP. A potential therapeutic target: The role of neutrophils in the central nervous system. Brain Behav Immun Health 2023; 33:100688. [PMID: 37767236 PMCID: PMC10520304 DOI: 10.1016/j.bbih.2023.100688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 09/14/2023] [Accepted: 09/16/2023] [Indexed: 09/29/2023] Open
Abstract
Neutrophils play a critical role in immune defense as the first recruited and most abundant leukocytes in the innate immune system. As such, regulation of neutrophil effector functions have strong implications on immunity. These cells display a wide heterogeneity of function, including both inflammatory and immunomodulatory roles. Neutrophils commonly infiltrate the central nervous system (CNS) in response to varied pathological conditions. There is still little understanding of the role these cells play in the CNS in such conditions. In the present review, we will summarize what is known of neutrophil's role in cancer and Alzheimer's disease (AD), with a focus on highlighting the gaps in our understanding.
Collapse
Affiliation(s)
- Isabel Salken
- College of Arts and Science, University of Virginia, USA
| | | | | |
Collapse
|
23
|
Wan X, Chou WK, Brynildsen MP. Amino acids can deplete ATP and impair nitric oxide detoxification by Escherichia coli. Free Radic Biol Med 2023; 205:90-99. [PMID: 37253411 DOI: 10.1016/j.freeradbiomed.2023.05.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/18/2023] [Accepted: 05/15/2023] [Indexed: 06/01/2023]
Abstract
Nitric oxide (·NO) is a prevalent antimicrobial that is known to damage iron-containing enzymes in amino acid (AA) biosynthesis pathways. With Escherichia coli, ·NO is detoxified in aerobic environments by Hmp, which is an enzyme that is synthesized de novo in response to ·NO. With this knowledgebase, it is expected that the availability of AAs in the extracellular environment would enhance ·NO detoxification, because AAs would foster translation of Hmp. However, we observed that ·NO detoxification by E. coli was far slower in populations grown and treated in the presence of AAs (AA+) in comparison to those grown and stressed in the absence of AAs (AA-). Further experiments revealed that AA+ populations had difficulty translating proteins under ·NO stress, and that ·NO activated the stringent response in AA+ populations. Additional work revealed significant ATP depletion in ·NO-stressed AA+ cultures that far exceeded that of ·NO-stressed AA- populations. Transcription, translation, and RelA were not found to be significant contributors to the ATP depletion observed, whereas AA import was implicated as a significant ATP consumption pathway. Alleviating ATP depletion while maintaining access to AAs partially restored ·NO detoxification, which suggested that ATP depletion contributed to the translational difficulties observed in ·NO-stressed AA+ populations. These data reveal an unexpected interaction within the ·NO response network of E. coli that stimulates a stringent response by RelA in conditions where AAs are plentiful.
Collapse
Affiliation(s)
- Xuanqing Wan
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, 08544, USA
| | - Wen Kang Chou
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, 08544, USA
| | - Mark P Brynildsen
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, 08544, USA.
| |
Collapse
|
24
|
Zawrotniak M, Juszczak M, Rapała-Kozik M. Release of neutrophil extracellular traps in response to Candida albicans yeast, as a secondary defense mechanism activated by phagocytosis. Yeast 2023; 40:349-359. [PMID: 36737224 DOI: 10.1002/yea.3842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/24/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023] Open
Abstract
Candida albicans is one of the main pathogens responsible for the development of difficult-to-fight fungal infections called candidiasis. Neutrophils are the major effector cells involved in the eradication of fungal pathogens. This group of immune cells uses several mechanisms that enable the rapid neutralization of pathogens. The most frequently identified mechanisms are phagocytosis and the release of neutrophil extracellular traps (NETs). The mechanism for selecting the type of neutrophil immune response is still unknown. In our study, we analyzed the relationship between the activation of phagocytosis and netosis. We detected the presence of two neutrophil populations characterized by different response patterns to contact with C. albicans blastospores. The first neutrophil population showed an increased ability to rapidly release NETs without prior internalization of the pathogen. In the second population, the netosis process was inherently associated with phagocytosis. Differences between populations also referred to the production of reactive oxygen species. Our results suggest that neutrophils use different strategies to fight C. albicans and, contrary to previous reports, these mechanisms are not mutually exclusive.
Collapse
Affiliation(s)
- Marcin Zawrotniak
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Magdalena Juszczak
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Maria Rapała-Kozik
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
25
|
Criado M, Pérez V, Arteche-Villasol N, Elguezabal N, Molina E, Benavides J, Gutiérrez-Expósito D. Evaluation of the innate immune response of caprine neutrophils against Mycobacterium avium subspecies paratuberculosis in vitro. Vet Res 2023; 54:61. [PMID: 37464437 DOI: 10.1186/s13567-023-01193-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 05/22/2023] [Indexed: 07/20/2023] Open
Abstract
Neutrophils constitute an essential component of the innate immune response, readily killing most bacteria through phagocytosis, degranulation, and the release of neutrophil extracellular traps (NETs) among other mechanisms. These cells play an unclear role in mycobacterial infections such as Mycobacterium avium subspecies paratuberculosis (Map), the etiological agent of paratuberculosis, and its response is particularly understudied in ruminants. Herein, a wide set of techniques were adapted, or newly developed, to study the in vitro response of caprine neutrophils after Map infection. Immunofluorescence was used to demonstrate, simultaneously, chemotaxis, phagocytosis, degranulation, and NETs. The quantification of neutrophil phagocytic activity against Map at a 1:10 multiplicity of infection (MOI), through flow cytometry, showed values that varied from 4.54 to 5.63% of phagocyting neutrophils. By immunofluorescence, a 73.3 ± 14.5% of the fields showed NETs, and the mean release of DNA, attributable to NETosis, calculated through a fluorometric method, was 16.2 ± 3.5%. In addition, the RNA expression of TGF-β, TNF and IL-1β cytokines, measured through reverse transcription qPCR, was significantly higher in the two latter. Overall, neutrophil response was proportional to the number of bacteria. This work confirms that the simultaneous study of several neutrophil mechanisms, and the combination of different methodologies, are essential to reach a comprehensive understanding of neutrophil response against pathogens, demonstrates that, in vitro, caprine neutrophils display a strong innate response against Map, using their entire repertoire of effector functions, and sets the basis for further in vitro and in vivo studies on the role of neutrophils in paratuberculosis.
Collapse
Affiliation(s)
- Miguel Criado
- Departamento de Sanidad Animal, Instituto de Ganadería de Montaña (IGM) CSIC-ULE, Grulleros, León, Spain.
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071, León, Spain.
| | - Valentín Pérez
- Departamento de Sanidad Animal, Instituto de Ganadería de Montaña (IGM) CSIC-ULE, Grulleros, León, Spain
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071, León, Spain
| | - Noive Arteche-Villasol
- Departamento de Sanidad Animal, Instituto de Ganadería de Montaña (IGM) CSIC-ULE, Grulleros, León, Spain
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071, León, Spain
| | - Natalia Elguezabal
- Departamento de Sanidad Animal, NEIKER-BRTA, Instituto Vasco de Investigación y Desarrollo Agrario, 48160, Derio, Vizcaya, Spain
| | - Elena Molina
- Departamento de Sanidad Animal, NEIKER-BRTA, Instituto Vasco de Investigación y Desarrollo Agrario, 48160, Derio, Vizcaya, Spain
| | - Julio Benavides
- Departamento de Sanidad Animal, Instituto de Ganadería de Montaña (IGM) CSIC-ULE, Grulleros, León, Spain
| | - Daniel Gutiérrez-Expósito
- Departamento de Sanidad Animal, Instituto de Ganadería de Montaña (IGM) CSIC-ULE, Grulleros, León, Spain
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071, León, Spain
| |
Collapse
|
26
|
Mankan AK, Czajka-Francuz P, Prendes M, Ramanan S, Koziej M, Vidal L, Saini KS. Intracellular DNA sensing by neutrophils and amplification of the innate immune response. Front Immunol 2023; 14:1208137. [PMID: 37483598 PMCID: PMC10361817 DOI: 10.3389/fimmu.2023.1208137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/13/2023] [Indexed: 07/25/2023] Open
Abstract
As the first responders, neutrophils lead the innate immune response to infectious pathogens and inflammation inducing agents. The well-established pathogen neutralizing strategies employed by neutrophils are phagocytosis, the action of microbicide granules, the production of ROS, and the secretion of neutrophil extracellular traps (NETs). Only recently, the ability of neutrophils to sense and respond to pathogen-associated molecular patterns is being appreciated. This review brings together the current information about the intracellular recognition of DNA by neutrophils and proposes models of signal amplification in immune response. Finally, the clinical relevance of DNA sensing by neutrophils in infectious and non-infectious diseases including malignancy are also discussed.
Collapse
Affiliation(s)
| | | | - Maria Prendes
- Labcorp Drug Development Inc., Princeton, NJ, United States
| | - Sriram Ramanan
- Labcorp Drug Development Inc., Princeton, NJ, United States
| | | | | | - Kamal S. Saini
- Fortrea, Inc., Durham, NC, United States
- Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| |
Collapse
|
27
|
Omar M, Abdelal H. NETosis in Parasitic Infections: A Puzzle That Remains Unsolved. Int J Mol Sci 2023; 24:ijms24108975. [PMID: 37240321 DOI: 10.3390/ijms24108975] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/06/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Neutrophils are the key players in the innate immune system, being weaponized with numerous strategies to eliminate pathogens. The production of extracellular traps is one of the effector mechanisms operated by neutrophils in a process called NETosis. Neutrophil extracellular traps (NETs) are complex webs of extracellular DNA studded with histones and cytoplasmic granular proteins. Since their first description in 2004, NETs have been widely investigated in different infectious processes. Bacteria, viruses, and fungi have been shown to induce the generation of NETs. Knowledge is only beginning to emerge about the participation of DNA webs in the host's battle against parasitic infections. Referring to helminthic infections, we ought to look beyond the scope of confining the roles of NETs solely to parasitic ensnarement or immobilization. Hence, this review provides detailed insights into the less-explored activities of NETs against invading helminths. In addition, most of the studies that have addressed the implications of NETs in protozoan infections have chiefly focused on their protective side, either through trapping or killing. Challenging this belief, we propose several limitations regarding protozoan-NETs interaction. One of many is the duality in the functional responses of NETs, in which both the positive and pathological aspects seem to be closely intertwined.
Collapse
Affiliation(s)
- Marwa Omar
- Department of Medical Parasitology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Heba Abdelal
- LIS: Cross-National Data Center, Maison des Sciences Humaines, Esch-Belval, L-4366 Luxembourg, Luxembourg
| |
Collapse
|
28
|
Adrover JM, McDowell SAC, He XY, Quail DF, Egeblad M. NETworking with cancer: The bidirectional interplay between cancer and neutrophil extracellular traps. Cancer Cell 2023; 41:505-526. [PMID: 36827980 DOI: 10.1016/j.ccell.2023.02.001] [Citation(s) in RCA: 120] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/09/2023] [Accepted: 02/01/2023] [Indexed: 02/25/2023]
Abstract
Neutrophils are major effectors and regulators of the immune system. They play critical roles not only in the eradication of pathogens but also in cancer initiation and progression. Conversely, the presence of cancer affects neutrophil activity, maturation, and lifespan. By promoting or repressing key neutrophil functions, cancer cells co-opt neutrophil biology to their advantage. This co-opting includes hijacking one of neutrophils' most striking pathogen defense mechanisms: the formation of neutrophil extracellular traps (NETs). NETs are web-like filamentous extracellular structures of DNA, histones, and cytotoxic granule-derived proteins. Here, we discuss the bidirectional interplay by which cancer stimulates NET formation, and NETs in turn support disease progression. We review how vascular dysfunction and thrombosis caused by neutrophils and NETs underlie an elevated risk of death from cardiovascular events in cancer patients. Finally, we propose therapeutic strategies that may be effective in targeting NETs in the clinical setting.
Collapse
Affiliation(s)
- Jose M Adrover
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Sheri A C McDowell
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada; Department of Physiology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Xue-Yan He
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Daniela F Quail
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada; Department of Physiology, Faculty of Medicine, McGill University, Montreal, QC, Canada.
| | - Mikala Egeblad
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA.
| |
Collapse
|
29
|
Martinez-Sanz P, Laurent ARG, Slot E, Hoogenboezem M, Bąbała N, van Bruggen R, Rongvaux A, Flavell RA, Tytgat GAM, Franke K, Matlung HL, Kuijpers TW, Amsen D, Karrich JJ. Humanized MISTRG as a preclinical in vivo model to study human neutrophil-mediated immune processes. Front Immunol 2023; 14:1105103. [PMID: 36969261 PMCID: PMC10032520 DOI: 10.3389/fimmu.2023.1105103] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 02/22/2023] [Indexed: 03/11/2023] Open
Abstract
IntroductionMISTRG mice have been genetically modified to allow development of a human myeloid compartment from engrafted human CD34+ haemopoietic stem cells, making them particularly suited to study the human innate immune system in vivo. Here, we characterized the human neutrophil population in these mice to establish a model that can be used to study the biology and contribution in immune processes of these cells in vivo.Methods and resultsWe could isolate human bone marrow neutrophils from humanized MISTRG mice and confirmed that all neutrophil maturation stages from promyelocytes (CD11b–CD16–) to end-stage segmented cells (CD11b+CD16+) were present. We documented that these cells possessed normal functional properties, including degranulation, reactive oxygen species production, adhesion, and antibody-dependent cellular cytotoxicity towards antibody-opsonized tumor cells ex vivo. The acquisition of functional capacities positively correlated with the maturation state of the cell. We found that human neutrophils were retained in the bone marrow of humanized MISTRG mice during steady state. However, the mature segmented CD11b+CD16+ human neutrophils were released from the bone marrow in response to two well-established neutrophil-mobilizing agents (i.e., G-CSF and/or CXCR4 antagonist Plerixafor). Moreover, the neutrophil population in the humanized MISTRG mice actively reacted to thioglycolate-induced peritonitis and could infiltrate implanted human tumors, as shown by flow cytometry and fluorescent microscopy.DiscussionThese results show that functional human neutrophils are generated and can be studied in vivo using the humanized MISTRG mice, providing a model to study the various functions of neutrophils in inflammation and in tumors.
Collapse
Affiliation(s)
- Paula Martinez-Sanz
- Sanquin Research and Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- *Correspondence: Paula Martinez-Sanz, ; Julien J. Karrich, ; Derk Amsen,
| | - Adrien R. G. Laurent
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Edith Slot
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Mark Hoogenboezem
- Sanquin Research and Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Nikolina Bąbała
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Robin van Bruggen
- Sanquin Research and Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Anthony Rongvaux
- Department of Immunology, University of Washington, Seattle, WA, United States
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, WA, United States
| | - Richard A. Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, United States
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, United States
| | - Godelieve A. M. Tytgat
- Princess Maxima Center for Pediatric Oncology, Department of Pediatric Oncology, Utrecht, Netherlands
| | - Katka Franke
- Sanquin Research and Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Hanke L. Matlung
- Sanquin Research and Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Taco W. Kuijpers
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Rheumatology and Infectious Diseases, Emma Children's Hospital, Department of Pediatric Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Derk Amsen
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- *Correspondence: Paula Martinez-Sanz, ; Julien J. Karrich, ; Derk Amsen,
| | - Julien J. Karrich
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- *Correspondence: Paula Martinez-Sanz, ; Julien J. Karrich, ; Derk Amsen,
| |
Collapse
|
30
|
Ribeiro IMM, de Sousa VC, Melo ECS, Carvalho RDCVD, Santos MDSD, Neto JADON, Melo DSD, Teixeira LSDA, Citó AMDGL, Moura AKS, Arcanjo DDR, Carvalho FADA, Alves MMDM, Mendonça ILD. Antileishmania and immunomodulatory potential of cashew nut shell liquid and cardanol. Toxicol In Vitro 2023; 87:105524. [PMID: 36435415 DOI: 10.1016/j.tiv.2022.105524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/25/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022]
Abstract
Conventional treatments for leishmaniasis have caused serious adverse effects, poor tolerance, development of resistant strains. Natural products have been investigated as potential therapeutic alternatives. The cashew nut shell liquid (CNSL) is a natural source of phenolic compounds with several biological activities, where cardanol (CN) is considered one of the most important and promising compounds. This study aimed to evaluate antileishmanial, cytotoxic and immunomodulatory activities of CNSL and CN. Both showed antileishmanial potential, with IC50 for CNSL and CN against Leishmania infantum: 148.12 and 56.74 μg/mL; against Leishmania braziliensis: 85.71 and 64.28 μg/mL; against Leishmania major: 153.56 and 122.31 μg/mL, respectively. The mean cytotoxic concentrations (CC50) of CNSL and CN were 37.51 and 31.44 μg/mL, respectively. CNSL and CN significantly reduced the percentage of infected macrophages, with a selectivity index (SI) >20 for CN. CNSL and cardanol caused an increase in phagocytic capacity and lysosomal volume. Survival rates of Zophobas morio larvae at doses of 3; 30 and 300 mg/kg were: 85%, 75% and 60% in contact with CNSL and 85%, 60% and 40% in contact with CN, respectively. There was a significant difference between the survival curves of larvae when treated with CN, demonstrating a significant acute toxicity for this substance. Additional investigations are needed to evaluate these substances in the in vivo experimental infection model.
Collapse
Affiliation(s)
- Iuliana Marjory Martins Ribeiro
- Programa de Pós-Graduação em Tecnologias Aplicadas a Animais de Interesse Regional, Universidade Federal do Piauí, Teresina, Piauí, Brazil
| | - Valéria Carlos de Sousa
- Núcleo de Pesquisas em Plantas Medicinais, Universidade Federal do Piauí, Teresina, Piauí, Brazil
| | | | | | | | | | - Danielly Silva de Melo
- Núcleo de Pesquisas em Plantas Medicinais, Universidade Federal do Piauí, Teresina, Piauí, Brazil
| | | | | | | | - Daniel Dias Rufino Arcanjo
- Núcleo de Pesquisas em Plantas Medicinais, Universidade Federal do Piauí, Teresina, Piauí, Brazil; Departamento de Biofísica e Fisiologia, Universidade Federal do Piauí, Teresina, Piauí, Brazil.
| | | | - Michel Muálem de Moraes Alves
- Programa de Pós-Graduação em Tecnologias Aplicadas a Animais de Interesse Regional, Universidade Federal do Piauí, Teresina, Piauí, Brazil; Núcleo de Pesquisas em Plantas Medicinais, Universidade Federal do Piauí, Teresina, Piauí, Brazil; Departamento de Morfofisiologia Veterinária, Universidade Federal do Piauí, Teresina, Piauí, Brazil.
| | - Ivete Lopes de Mendonça
- Programa de Pós-Graduação em Tecnologias Aplicadas a Animais de Interesse Regional, Universidade Federal do Piauí, Teresina, Piauí, Brazil; Departamento de Clínica e Cirurgia Veterinária, Universidade Federal do Piauí, Teresina, Piauí, Brazil
| |
Collapse
|
31
|
Andersen G, Kahlenberg K, Krautwurst D, Somoza V. [6]-Gingerol Facilitates CXCL8 Secretion and ROS Production in Primary Human Neutrophils by Targeting the TRPV1 Channel. Mol Nutr Food Res 2023; 67:e2200434. [PMID: 36564924 DOI: 10.1002/mnfr.202200434] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 12/13/2022] [Indexed: 12/25/2022]
Abstract
SCOPE Clarifying the function of sensory active TRP (transient receptor potential) channels in non-sensory tissue is of growing interest, especially with regard to food ingredients in nutritionally relevant concentrations. The study hypothesizes the TRPV1 agonist [6]-gingerol to facilitate cellular immune responses of primary human neutrophils, after treatment with 50 nM, a concentration that can be reached in the circulation after habitual dietary intake. METHODS AND RESULTS qRT-PCR analyses reveal a high abundancy of TRP channel RNA expression in the types of primary leukocytes investigated, namely neutrophils, monocytes, NK cells, T cells, and B cells. Incubation of neutrophils with 50 nM of the known TRPV1 ligand [6]-gingerol led to increased surface expression of CD11b, CD66b, and the fMLF receptor FPR1, as shown by flow cytometry. Upon subsequent stimulation with fMLF, the neutrophils display an about 30% (p < 0.05) increase in CXCL8 secretion as well as in ROS production. Pharmacological inhibition of TRPV1 by trans-tert-butylcyclohexanol abolishes the [6]-gingerol induced effects. CONCLUSIONS The TRPV1 channel is functionally expressed in human neutrophils. Activation of the channel with [6]-gingerol as a food-derived ligand in nutritionally relevant concentrations leads to an enhanced responsiveness in the cells towards activating stimuli, thereby facilitating a canonical cellular immune response in human neutrophils.
Collapse
Affiliation(s)
- Gaby Andersen
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, 85354, Freising, Germany
| | - Kristin Kahlenberg
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, 85354, Freising, Germany
| | - Dietmar Krautwurst
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, 85354, Freising, Germany
| | - Veronika Somoza
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, 85354, Freising, Germany.,Chair of Nutritional Systems Biology at the Technical University of Munich, 85354, Freising, Germany.,Department of Nutritional and Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, 1090, Austria
| |
Collapse
|
32
|
Bai S, Wei Y, Liu R, Chen Y, Ma W, Wang M, Chen L, Luo Y, Du J. The role of transient receptor potential channels in metastasis. Biomed Pharmacother 2023; 158:114074. [PMID: 36493698 DOI: 10.1016/j.biopha.2022.114074] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 11/30/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Metastasis is the hallmark of failed tumor treatment and is typically associated with death due to cancer. Transient receptor potential (TRP) channels affect changes in intracellular calcium concentrations and participate at every stage of metastasis. Further, they increase the migratory ability of tumor cells, promote angiogenesis, regulate immune function, and promote the growth of tumor cells through changes in gene expression and function. In this review, we explore the potential mechanisms of action of TRP channels, summarize their role in tumor metastasis, compile inhibitors of TRP channels relevant in tumors, and discuss current challenges in research on TRP channels involved in tumor metastasis.
Collapse
Affiliation(s)
- Suwen Bai
- Longgang District People's Hospital of Shenzhen & The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Yuan Wei
- Longgang District People's Hospital of Shenzhen & The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Rong Liu
- School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China
| | - Yuhua Chen
- Longgang District People's Hospital of Shenzhen & The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Wanling Ma
- Longgang District People's Hospital of Shenzhen & The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Minghua Wang
- Longgang District People's Hospital of Shenzhen & The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Li Chen
- Department of obstetrics and gynecology, The Seventh Affiliated Hospital, Sun Yat-sen University, Zhenyuan Rd, Guangming Dist., Shenzhen, Guangdong 518107, China
| | - Yumei Luo
- Longgang District People's Hospital of Shenzhen & The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China.
| | - Juan Du
- Ciechanover Institute of Precision and Regenerative Medicine, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China.
| |
Collapse
|
33
|
Nun N, Joy A. Fabrication and Bioactivity of Peptide-Conjugated Biomaterial Tissue Engineering Constructs. Macromol Rapid Commun 2023; 44:e2200342. [PMID: 35822458 DOI: 10.1002/marc.202200342] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 06/22/2022] [Indexed: 01/11/2023]
Abstract
Tissue engineering combines materials engineering, cells and biochemical factors to improve, restore or replace various types of biological tissues. A nearly limitless combination of these strategies can be combined, providing a means to augment the function of a number of biological tissues such as skin tissue, neural tissue, bones, and cartilage. Compounds such as small molecule therapeutics, proteins, and even living cells have been incorporated into tissue engineering constructs to influence biological processes at the site of implantation. Peptides have been conjugated to tissue engineering constructs to circumvent limitations associated with conjugation of proteins or incorporation of cells. This review highlights various contemporary examples in which peptide conjugation is used to overcome the disadvantages associated with the inclusion of other bioactive compounds. This review covers several peptides that are commonly used in the literature as well as those that do not appear as frequently to provide a broad scope of the utility of the peptide conjugation technique for designing constructs capable of influencing the repair and regeneration of various bodily tissues. Additionally, a brief description of the construct fabrication techniques encountered in the covered examples and their advantages in various tissue engineering applications is provided.
Collapse
Affiliation(s)
- Nicholas Nun
- School of Polymer Science and Polymer Engineering, The University of Akron, Akron, OH, 44321, USA
| | - Abraham Joy
- School of Polymer Science and Polymer Engineering, The University of Akron, Akron, OH, 44321, USA
| |
Collapse
|
34
|
Soliman AM, Barreda DR. Acute Inflammation in Tissue Healing. Int J Mol Sci 2022; 24:ijms24010641. [PMID: 36614083 PMCID: PMC9820461 DOI: 10.3390/ijms24010641] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/19/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
There are well-established links between acute inflammation and successful tissue repair across evolution. Innate immune reactions contribute significantly to pathogen clearance and activation of subsequent reparative events. A network of molecular and cellular regulators supports antimicrobial and tissue repair functions throughout the healing process. A delicate balance must be achieved between protection and the potential for collateral tissue damage associated with overt inflammation. In this review, we summarize the contributions of key cellular and molecular components to the acute inflammatory process and the effective and timely transition toward activation of tissue repair mechanisms. We further discuss how the disruption of inflammatory responses ultimately results in chronic non-healing injuries.
Collapse
Affiliation(s)
- Amro M. Soliman
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Daniel R. Barreda
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Correspondence: ; Tel.: +1-(780)492-0375
| |
Collapse
|
35
|
Guan J. A Theoretical Model for Phagocytic Capacity of Phagocytes. ADVANCED THEORY AND SIMULATIONS 2022. [DOI: 10.1002/adts.202200710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Jingjiao Guan
- Department of Chemical and Biomedical Engineering FAMU‐FSU College of Engineering Florida State University 2525 Pottsdamer Street Tallahassee FL 32310‐2870 USA
| |
Collapse
|
36
|
Hosseinalizadeh H, Mahmoodpour M, Razaghi Bahabadi Z, Hamblin MR, Mirzaei H. Neutrophil mediated drug delivery for targeted glioblastoma therapy: A comprehensive review. Biomed Pharmacother 2022; 156:113841. [DOI: 10.1016/j.biopha.2022.113841] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 10/02/2022] [Accepted: 10/06/2022] [Indexed: 11/08/2022] Open
|
37
|
Alves-Soares R, de Oliveira HD, Campos DDCO, Gomes-Vasconcelos YDA, Ferreira-da-Silva FW, Silva-Alves KS, Coelho-de-Souza LN, Diniz LRL, Leal-Cardoso JH, Coelho-de-Souza AN. The Essential Oil of Hyptis crenata Inhibits the Increase in Secretion of Inflammatory Mediators. PLANTS (BASEL, SWITZERLAND) 2022; 11:3048. [PMID: 36432777 PMCID: PMC9695298 DOI: 10.3390/plants11223048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/03/2022] [Accepted: 11/03/2022] [Indexed: 06/16/2023]
Abstract
Background: Hyptis crenata is a plant of great ethnopharmacological importance widely distributed in South American countries. In Northeast Brazil, teas or infusions of its aerial parts are used in folk medicine to treat several acute and chronic inflammatory diseases. In a previous work we have demonstrated that the essential oil of H. crenata (EOHc) has an antiedematogenic effect. The aim of this work was to evaluate the effect of EOHc on cytokines secretion and cellular infiltration. Methods: Peritonitis and paw edema models induced by carrageenan were used to determine leucocyte count, myeloperoxidase (MPO) activity, nitrite, and cytokines secretion. Results: EOHc (10−300 mg/kg) significantly inhibited leucocyte migration and reduced the neutrophil count (control: 1.46 × 103 ± 0.031 × 103/mL) of the total leucocytes population in extracellular exudate (control: 2.14 × 103 ± 0.149 × 103/mL) by 15.00%, 43.29%, 65.52%, and 72.83% for the doses of 10, 30, 100, and 300 mg/kg EOHc, respectively (EC50: 24.15 mg/kg). EOHc (100 mg/kg) inhibited the increase in myeloperoxidase activity and completely blocked the increase in nitrite concentration induced by carrageenan. EOHc markedly reduced the pro-inflammatory cytokines (IL-6, MCP-1, IFN-γ, TNF-α, and IL-12p70) and increased IL-10, an anti-inflammatory cytokine (compared to control group, p < 0.05). Conclusions: This study demonstrates that EOHc has a long-lasting anti-inflammatory effect mediated through interference on MPO activity, and nitrite, and cytokines secretion. This effect, coupled with low EOHc toxicity, as far as results obtained in mice could be translated to humans, suggests that EOHc has great potentiality as a therapeutic agent.
Collapse
Affiliation(s)
- Rutyleia Alves-Soares
- Postgraduate Program in Physiological Sciences, Superior Institute of Biomedical Sciences (ISCB), Campus do Itaperi, State University of Ceara (UECE), Av. Silas Munguba 1700, Fortaleza CEP 60.714.903, Ceara, Brazil
| | - Hermógenes David de Oliveira
- Postgraduate Program in Biochemistry, Department of Biochemistry and Molecular Biology, Campus do Pici, Federal University of Ceara (UFC), Av. Humberto Monte, s/n Bloco 907, Fortaleza CEP 60440-990, Ceara, Brazil
| | - Dyély de Carvalho Oliveira Campos
- Postgraduate Program in Physiological Sciences, Superior Institute of Biomedical Sciences (ISCB), Campus do Itaperi, State University of Ceara (UECE), Av. Silas Munguba 1700, Fortaleza CEP 60.714.903, Ceara, Brazil
| | - Yuri de Abreu Gomes-Vasconcelos
- Postgraduate Program in Physiological Sciences, Superior Institute of Biomedical Sciences (ISCB), Campus do Itaperi, State University of Ceara (UECE), Av. Silas Munguba 1700, Fortaleza CEP 60.714.903, Ceara, Brazil
| | - Francisco Walber Ferreira-da-Silva
- Postgraduate Program in Physiological Sciences, Superior Institute of Biomedical Sciences (ISCB), Campus do Itaperi, State University of Ceara (UECE), Av. Silas Munguba 1700, Fortaleza CEP 60.714.903, Ceara, Brazil
| | - Kerly Shamyra Silva-Alves
- Postgraduate Program in Physiological Sciences, Superior Institute of Biomedical Sciences (ISCB), Campus do Itaperi, State University of Ceara (UECE), Av. Silas Munguba 1700, Fortaleza CEP 60.714.903, Ceara, Brazil
| | - Lianna Noronha Coelho-de-Souza
- Graduate Program of Medicine, Center for Health Sciences (CCS), Fortaleza University (UNIFOR), Av. Washington Soares 1321, Edson Queiroz, Fortaleza CEP 60811-905, Ceara, Brazil
| | - Lúcio Ricardo Leite Diniz
- Researcher at the National Institute of the Semiarid Region (INSA), Av. Francisco Lopes de Almeida, s/n Serrotão, Campina Grande CEP 58434-70, Paraiba, Brazil
| | - José Henrique Leal-Cardoso
- Postgraduate Program in Physiological Sciences, Superior Institute of Biomedical Sciences (ISCB), Campus do Itaperi, State University of Ceara (UECE), Av. Silas Munguba 1700, Fortaleza CEP 60.714.903, Ceara, Brazil
| | - Andrelina Noronha Coelho-de-Souza
- Postgraduate Program in Physiological Sciences, Superior Institute of Biomedical Sciences (ISCB), Campus do Itaperi, State University of Ceara (UECE), Av. Silas Munguba 1700, Fortaleza CEP 60.714.903, Ceara, Brazil
| |
Collapse
|
38
|
Pérez-Figueroa E, Álvarez-Carrasco P, Ortega E. Crosslinking of membrane CD13 in human neutrophils mediates phagocytosis and production of reactive oxygen species, neutrophil extracellular traps and proinflammatory cytokines. Front Immunol 2022; 13:994496. [PMID: 36439182 PMCID: PMC9686367 DOI: 10.3389/fimmu.2022.994496] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/19/2022] [Indexed: 09/08/2023] Open
Abstract
Aminopeptidase N, or CD13, is a cell membrane ectopeptidase highly expressed in myeloid cells. Through its enzymatic activity, CD13 regulates the activity of several bioactive peptides, such as endorphins and enkephalins, chemotactic peptides like MCP-1 and IL-8, angiotensin III, bradikinin, etc. In recent years, it has been appreciated that independently of its peptidase activity, CD13 can activate signal transduction pathways and mediate effector functions such as phagocytosis and cytokine secretion in monocytes and macrophages. Although neutrophils are known to express CD13 on its membrane, it is currently unknown if CD13 can mediate effector functions in these cells. Here, we show that in human neutrophils CD13 can mediate phagocytosis, which is dependent on a signaling pathway that involves Syk, and PI3-K. Phagocytosis mediated by CD13 is associated with production of reactive oxygen species (ROS). The level of phagocytosis and ROS production mediated by CD13 are similar to those through FcγRIII (CD16b), a widely studied receptor of human neutrophils. Also, CD13 ligation induces the release of neutrophil extracellular traps (NETs) as well as cytokine secretion from neutrophils. These results support the hypothesis that CD13 is a membrane receptor able to activate effector functions in human neutrophils.
Collapse
Affiliation(s)
| | | | - Enrique Ortega
- Department of Immunology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de Mexico, Mexico
| |
Collapse
|
39
|
Immunosenescence, Inflammaging, and Lung Senescence in Asthma in the Elderly. Biomolecules 2022; 12:biom12101456. [PMID: 36291665 PMCID: PMC9599177 DOI: 10.3390/biom12101456] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/02/2022] [Accepted: 10/04/2022] [Indexed: 11/24/2022] Open
Abstract
Prevalence of asthma in older adults is growing along with increasing global life expectancy. Due to poor clinical consequences such as high mortality, advancement in understanding the pathophysiology of asthma in older patients has been sought to provide prompt treatment for them. Age-related alterations of functions in the immune system and lung parenchyma occur throughout life. Alterations with advancing age are promoted by various stimuli, including pathobionts, fungi, viruses, pollutants, and damage-associated molecular patterns derived from impaired cells, abandoned cell debris, and senescent cells. Age-related changes in the innate and adaptive immune response, termed immunosenescence, includes impairment of phagocytosis and antigen presentation, enhancement of proinflammatory mediator generation, and production of senescence-associated secretory phenotype. Immnunosenescence could promote inflammaging (chronic low-grade inflammation) and contribute to late-onset adult asthma and asthma in the elderly, along with age-related pulmonary disease, such as chronic obstructive pulmonary disease and pulmonary fibrosis, due to lung parenchyma senescence. Aged patients with asthma exhibit local and systemic type 2 and non-type 2 inflammation, associated with clinical manifestations. Here, we discuss immunosenescence’s contribution to the immune response and the combination of type 2 inflammation and inflammaging in asthma in the elderly and present an overview of age-related features in the immune system and lung structure.
Collapse
|
40
|
Belchamber KBR, Thein OS, Hazeldine J, Grudzinska FS, Faniyi AA, Hughes MJ, Jasper AE, Yip KP, Crowley LE, Lugg ST, Sapey E, Parekh D, Thickett DR, Scott A. Dysregulated Neutrophil Phenotype and Function in Hospitalised Non-ICU COVID-19 Pneumonia. Cells 2022; 11:2901. [PMID: 36139476 PMCID: PMC9496854 DOI: 10.3390/cells11182901] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/09/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Rationale: Infection with the SARS-CoV2 virus is associated with elevated neutrophil counts. Evidence of neutrophil dysfunction in COVID-19 is based on transcriptomics or single functional assays. Cell functions are interwoven pathways, and understanding the effect across the spectrum of neutrophil function may identify therapeutic targets. Objectives: Examine neutrophil phenotype and function in 41 hospitalised, non-ICU COVID-19 patients versus 23 age-matched controls (AMC) and 26 community acquired pneumonia patients (CAP). Methods: Isolated neutrophils underwent ex vivo analyses for migration, bacterial phagocytosis, ROS generation, NETosis and receptor expression. Circulating DNAse 1 activity, levels of cfDNA, MPO, VEGF, IL-6 and sTNFRI were measured and correlated to clinical outcome. Serial sampling on day three to five post hospitalization were also measured. The effect of ex vivo PI3K inhibition was measured in a further cohort of 18 COVID-19 patients. Results: Compared to AMC and CAP, COVID-19 neutrophils demonstrated elevated transmigration (p = 0.0397) and NETosis (p = 0.0332), and impaired phagocytosis (p = 0.0036) associated with impaired ROS generation (p < 0.0001). The percentage of CD54+ neutrophils (p < 0.001) was significantly increased, while surface expression of CD11b (p = 0.0014) and PD-L1 (p = 0.006) were significantly decreased in COVID-19. COVID-19 and CAP patients showed increased systemic markers of NETosis including increased cfDNA (p = 0.0396) and impaired DNAse activity (p < 0.0001). The ex vivo inhibition of PI3K γ and δ reduced NET release by COVID-19 neutrophils (p = 0.0129). Conclusions: COVID-19 is associated with neutrophil dysfunction across all main effector functions, with altered phenotype, elevated migration and NETosis, and impaired antimicrobial responses. These changes highlight that targeting neutrophil function may help modulate COVID-19 severity.
Collapse
Affiliation(s)
- Kylie B. R. Belchamber
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
| | - Onn S. Thein
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
| | - Jon Hazeldine
- National Institute for Health Research Surgical Reconstruction and Microbiology Research Centre, Queen Elizabeth Hospital Birmingham, Birmingham B15 2TH, UK
| | - Frances S. Grudzinska
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
| | - Aduragbemi A. Faniyi
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
| | - Michael J. Hughes
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
| | - Alice E. Jasper
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
| | - Kay Por Yip
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
| | - Louise E. Crowley
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
| | - Sebastian T. Lugg
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
| | - Elizabeth Sapey
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
- PIONEER HDR-UK Hub in Acute Care, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
| | - Dhruv Parekh
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
- NIHR Clinical Research Facility, University Hospitals Birmingham NHS Foundation Trust, Edgbaston, Birmingham B12 2GW, UK
| | - David R. Thickett
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
| | - Aaron Scott
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
| |
Collapse
|
41
|
Single-cell RNA-seq of primary bone marrow neutrophils from female and male adult mice. Sci Data 2022; 9:442. [PMID: 35871169 PMCID: PMC9308797 DOI: 10.1038/s41597-022-01544-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/08/2022] [Indexed: 01/21/2023] Open
Abstract
Widespread sex-dimorphism is observed in the mammalian immune system. Consistently, studies have reported sex differences in the transcriptome of immune cells at the bulk level, including neutrophils. Neutrophils are the most abundant cell type in human blood, and they are key components of the innate immune system as they form a first line of defense against pathogens. Neutrophils are produced in the bone marrow, and differentiation and maturation produce distinct neutrophil subpopulations. Thus, single-cell resolution studies are crucial to decipher the biological significance of neutrophil heterogeneity. However, since neutrophils are very RNA-poor, single-cell profiling of these cells has been technically challenging. Here, we generated a single-cell RNA-seq dataset of primary neutrophils from adult female and male mouse bone marrow. After stringent quality control, we found that previously characterized neutrophil subpopulations can be detected in both sexes. Additionally, we confirmed that canonical sex-linked markers are differentially expressed between female and male cells across neutrophil subpopulations. This dataset provides a groundwork for comparative studies on the lifelong transcriptional sexual dimorphism of neutrophils.
Collapse
|
42
|
Moniot A, Braux J, Siboni R, Guillaume C, Audonnet S, Allart-Simon I, Sapi J, Tirouvanziam R, Gérard S, Gangloff SC, Velard F. Inhibition of Recruitment and Activation of Neutrophils by Pyridazinone-Scaffold-Based Compounds. Int J Mol Sci 2022; 23:ijms23137226. [PMID: 35806233 PMCID: PMC9266889 DOI: 10.3390/ijms23137226] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/25/2022] [Accepted: 06/26/2022] [Indexed: 12/07/2022] Open
Abstract
In inflammatory diseases, polymorphonuclear neutrophils (PMNs) are known to produce elevated levels of pro-inflammatory cytokines and proteases. To limit ensuing exacerbated cell responses and tissue damage, novel therapeutic agents are sought. 4aa and 4ba, two pyridazinone-scaffold-based phosphodiesterase-IV inhibitors are compared in vitro to zardaverine for their ability to: (1) modulate production of pro-inflammatory mediators, reactive oxygen species (ROS), and phagocytosis; (2) modulate degranulation by PMNs after transepithelial lung migration. Compound 4ba and zardaverine were tested in vivo for their ability to limit tissue recruitment of PMNs in a murine air pouch model. In vitro treatment of lipopolysaccharide-stimulated PMNs with compounds 4aa and 4ba inhibited the release of interleukin-8, tumor necrosis factor-α, and matrix metalloproteinase-9. PMNs phagocytic ability, but not ROS production, was reduced following treatment. Using a lung inflammation model, we proved that PMNs transmigration led to reduced expression of the CD16 phagocytic receptor, which was significantly blunted after treatment with compound 4ba or zardaverine. Using the murine air pouch model, LPS-induced PMNs recruitment was significantly decreased upon addition of compound 4ba or zardaverine. Our data suggest that new pyridazinone derivatives have therapeutic potential in inflammatory diseases by limiting tissue recruitment and activation of PMNs.
Collapse
Affiliation(s)
- Aurélie Moniot
- Université de Reims Champagne-Ardenne, EA 4691 BIOS, 51 Rue Cognacq Jay, 51100 Reims, France; (A.M.); (J.B.); (R.S.); (C.G.); (S.C.G.)
| | - Julien Braux
- Université de Reims Champagne-Ardenne, EA 4691 BIOS, 51 Rue Cognacq Jay, 51100 Reims, France; (A.M.); (J.B.); (R.S.); (C.G.); (S.C.G.)
| | - Renaud Siboni
- Université de Reims Champagne-Ardenne, EA 4691 BIOS, 51 Rue Cognacq Jay, 51100 Reims, France; (A.M.); (J.B.); (R.S.); (C.G.); (S.C.G.)
| | - Christine Guillaume
- Université de Reims Champagne-Ardenne, EA 4691 BIOS, 51 Rue Cognacq Jay, 51100 Reims, France; (A.M.); (J.B.); (R.S.); (C.G.); (S.C.G.)
| | - Sandra Audonnet
- Université de Reims Champagne-Ardenne, URCACyt, 51 Rue Cognacq Jay, 51100 Reims, France;
| | - Ingrid Allart-Simon
- Université de Reims Champagne-Ardenne, UMR CNRS 7312 ICMR, 51 Rue Cognacq Jay, 51100 Reims, France; (I.A.-S.); (J.S.); (S.G.)
| | - Janos Sapi
- Université de Reims Champagne-Ardenne, UMR CNRS 7312 ICMR, 51 Rue Cognacq Jay, 51100 Reims, France; (I.A.-S.); (J.S.); (S.G.)
| | - Rabindra Tirouvanziam
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA;
- Center for CF and Airways Disease Research, Children’s Healthcare of Atlanta, 2015 Uppergate Road, Atlanta, GA 30322, USA
| | - Stéphane Gérard
- Université de Reims Champagne-Ardenne, UMR CNRS 7312 ICMR, 51 Rue Cognacq Jay, 51100 Reims, France; (I.A.-S.); (J.S.); (S.G.)
| | - Sophie C. Gangloff
- Université de Reims Champagne-Ardenne, EA 4691 BIOS, 51 Rue Cognacq Jay, 51100 Reims, France; (A.M.); (J.B.); (R.S.); (C.G.); (S.C.G.)
| | - Frédéric Velard
- Université de Reims Champagne-Ardenne, EA 4691 BIOS, 51 Rue Cognacq Jay, 51100 Reims, France; (A.M.); (J.B.); (R.S.); (C.G.); (S.C.G.)
- Correspondence: ; Tel.: +33-3-26-91-80-10
| |
Collapse
|
43
|
Piccolo EB, Thorp EB, Sumagin R. Functional implications of neutrophil metabolism during ischemic tissue repair. Curr Opin Pharmacol 2022; 63:102191. [PMID: 35276496 PMCID: PMC8995387 DOI: 10.1016/j.coph.2022.102191] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/10/2022] [Accepted: 01/17/2022] [Indexed: 12/11/2022]
Abstract
Immune cell mobilization and their accumulation in the extravascular space is a key consequence of tissue injury. Maladaptive trafficking and immune activation following reperfusion of ischemic tissue can exacerbate tissue repair. After ischemic injury such as myocardial infarction (MI), PMNs are the first cells to arrive at the sites of insult and their response is critical for the sequential progression of ischemia from inflammation to resolution and finally to tissue repair. However, PMN-induced inflammation can also be detrimental to cardiac function and ultimately lead to heart failure. In this review, we highlight the role of PMNs during key cellular and molecular events of ischemic heart failure. We address new research on PMN metabolism, and how this orchestrates diverse functions such as PMN chemotaxis, degranulation, and phagocytosis. Particular focus is given to PMN metabolism regulation by mitochondrial function and mTOR kinase activity.
Collapse
Affiliation(s)
- Enzo B Piccolo
- Department of Pathology, Northwestern University Feinberg School of Medicine, 300 East Superior St, Chicago, IL, 60611, USA
| | - Edward B Thorp
- Department of Pathology, Northwestern University Feinberg School of Medicine, 300 East Superior St, Chicago, IL, 60611, USA.
| | - Ronen Sumagin
- Department of Pathology, Northwestern University Feinberg School of Medicine, 300 East Superior St, Chicago, IL, 60611, USA.
| |
Collapse
|
44
|
Blufstein A, Behm C, Kubin B, Gahn J, Moritz A, Rausch‐Fan X, Andrukhov O. Anti-apoptotic effects of human gingival mesenchymal stromal cells on polymorphonuclear leucocytes. Oral Dis 2022; 28:777-785. [PMID: 33386669 PMCID: PMC9290793 DOI: 10.1111/odi.13768] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 12/14/2020] [Accepted: 12/28/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Polymorphonuclear leucocytes (PMNs) constitute the first line of host defence and are crucial in maintaining periodontal health. Their survival and function are modulated by mesenchymal stromal cells (MSCs) from different origin. Gingival MSCs (GMSCs) play an important role in maintaining oral health and in the initial inflammatory response. The present study aimed to investigate the effects of GMSCs on PMNs apoptosis and reactive oxygen species (ROS) production. METHODS PMNs were either directly incubated with untreated, interleukin (IL)-1β- or tumour necrosis factor (TNF)-α-treated GMSCs or stimulated with their conditioned media. Resulting ROS production was evaluated by dichlorofluorescin diacetate staining, whereas PMNs apoptosis was assessed by Annexin V staining, followed by flow cytometry analysis. RESULTS While conditioned media of untreated and TNF-α-treated GMSCs did not affect apoptosis of PMNs, it was significantly delayed by conditioned media of GMSCs treated with IL-1β. In direct co-culture, GMSCs exerted anti-apoptotic effects on PMNs independently of the previous stimulation. However, the strongest impact was observed by IL-1β-treated GMSCs. ROS production of PMNs was not influenced by GMSCs or their conditioned media. CONCLUSION This study demonstrates for the first time the immunomodulatory properties of GMSCs towards PMNs, revealing that IL-1β enhances anti-apoptotic effects of GMSCs.
Collapse
Affiliation(s)
- Alice Blufstein
- Department of Conservative Dentistry and PeriodontologyUniversity Clinic of DentistryMedical University of ViennaViennaAustria
| | - Christian Behm
- Department of Conservative Dentistry and PeriodontologyUniversity Clinic of DentistryMedical University of ViennaViennaAustria
| | - Barbara Kubin
- Department of Conservative Dentistry and PeriodontologyUniversity Clinic of DentistryMedical University of ViennaViennaAustria
| | - Johannes Gahn
- Department of Conservative Dentistry and PeriodontologyUniversity Clinic of DentistryMedical University of ViennaViennaAustria
| | - Andreas Moritz
- Department of Conservative Dentistry and PeriodontologyUniversity Clinic of DentistryMedical University of ViennaViennaAustria
| | - Xiaohui Rausch‐Fan
- Department of Conservative Dentistry and PeriodontologyUniversity Clinic of DentistryMedical University of ViennaViennaAustria
| | - Oleh Andrukhov
- Department of Conservative Dentistry and PeriodontologyUniversity Clinic of DentistryMedical University of ViennaViennaAustria
| |
Collapse
|
45
|
Sanches RCO, Mambelli F, Oliveira SC. Neutrophils and schistosomiasis: a missing piece in pathology. Parasite Immunol 2022; 44:e12916. [PMID: 35332932 DOI: 10.1111/pim.12916] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/10/2022] [Accepted: 03/18/2022] [Indexed: 12/01/2022]
Abstract
Schistosomiasis is a chronic human parasitic disease that causes serious health problems worldwide. The disease-associated liver pathology is one of the hallmarks of infections by S. mansoni and S. japonicum, and is accountable for the debilitating condition found in infected patients. In the past few years, investigative studies have highlighted the key role played by neutrophils and the influence of inflammasome signaling pathway in different pathological conditions. However, it is noteworthy that the study of inflammasome activation in neutrophils has been overlooked by reports concerning macrophages and monocytes. This interplay between neutrophils and inflammasomes is much more poorly investigated during schistosomiasis. Herein we reviewed the role of neutrophils during schistosomiasis and addressed the potential connection between these cells and inflammasome activation in this context.
Collapse
Affiliation(s)
- Rodrigo C O Sanches
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Fábio Mambelli
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Sergio C Oliveira
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), CNPq MCT, Salvador, Brazil
| |
Collapse
|
46
|
Neutrophils in Extravascular Body Fluids: Cytological-Energy Analysis Enables Rapid, Reliable and Inexpensive Detection of Purulent Inflammation and Tissue Damage. Life (Basel) 2022; 12:life12020160. [PMID: 35207447 PMCID: PMC8877237 DOI: 10.3390/life12020160] [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: 12/11/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 11/17/2022] Open
Abstract
The simultaneous cytological and metabolic investigation of various extravascular body fluids (EBFs) provides clinically relevant information about the type and intensity of the immune response in particular organ systems. The oxidative burst of professional phagocytes with the concomitant production of reactive oxygen species consumes a large amount of oxygen and is the cause of switch to the development of anaerobic metabolism. We assessed the relationships between percentages of neutrophils, aerobic and anaerobic metabolism, and tissue damage via the determination of aspartate aminotransferase catalytic activities (AST) in cerebrospinal fluid (CSF), pleural effusions (PE), abdominal effusions (AE), and synovial fluids (SF). EBFs with 0.0–20.0% neutrophils: 83.0% aerobic and 1.3% strongly anaerobic cases with median of AST = 13.8 IU/L in CSF; 68.0% aerobic and 9.0% strongly anaerobic cases with median of AST = 20.4 IU/L in PE; 77.5% aerobic and 10.5% strongly anaerobic cases with median of AST = 18.0 IU/L in AE; 64.1% aerobic and 7.7% strongly anaerobic cases with median of AST = 13.8 IU/L in SF. EBFs with 80.0–100.0% neutrophils: 4.2% aerobic and 73.7% strongly anaerobic cases with median of AST = 19.2 IU/L in CSF; 7.4% aerobic and 77.3% strongly anaerobic cases with median of AST = 145.2 IU/L in PE; 11.8% aerobic and 73.7% strongly anaerobic cases with median of AST = 61.8 IU/L in AE; 25.5% aerobic and 38.2% strongly anaerobic cases with median of AST = 37.2 IU/L in SF. The significant presence of neutrophils, concomitant strong anaerobic metabolism, and elevated AST in various EBFs are reliable signs of damaging purulent inflammation.
Collapse
|
47
|
Rubey KM, Mukhitov AR, Nong J, Wu J, Krymskaya VP, Myerson JW, Worthen GS, Brenner JS. Nanoparticle-Induced Augmentation of Neutrophils' Phagocytosis of Bacteria. Front Pharmacol 2022; 13:923814. [PMID: 35860017 PMCID: PMC9289463 DOI: 10.3389/fphar.2022.923814] [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: 04/19/2022] [Accepted: 05/31/2022] [Indexed: 11/21/2022] Open
Abstract
Despite the power of antibiotics, bacterial infections remain a major killer, due to antibiotic resistance and hosts with dysregulated immune systems. We and others have been developing drug-loaded nanoparticles that home to the sites of infection and inflammation via engineered tropism for neutrophils, the first-responder leukocytes in bacterial infections. Here, we examined how a member of a broad class of neutrophil-tropic nanoparticles affects neutrophil behavior, specifically questioning whether the nanoparticles attenuate an important function, bacterial phagocytosis. We found these nanoparticles actually augment phagocytosis of non-opsonized bacteria, increasing it by ∼50%. We showed this augmentation of phagocytosis is likely co-opting an evolved response, as opsonized bacteria also augment phagocytosis of non-opsonized bacteria. Enhancing phagocytosis of non-opsonized bacteria may prove particularly beneficial in two clinical situations: in hypocomplementemic patients (meaning low levels of the main bacterial opsonins, complement proteins, seen in conditions such as neonatal sepsis and liver failure) or for bacteria that are largely resistant to complement opsonization (e.g., Neisseria). Additionally, we observe that; 1) prior treatment with bacteria augments neutrophil uptake of neutrophil-tropic nanoparticles; 2) neutrophil-tropic nanoparticles colocalize with bacteria inside of neutrophils. The observation that neutrophil-tropic nanoparticles enhance neutrophil phagocytosis and localize with bacteria inside neutrophils suggests that these nanoparticles will serve as useful carriers for drugs to ameliorate bacterial diseases.
Collapse
Affiliation(s)
- Kathryn M Rubey
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Alexander R Mukhitov
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Jia Nong
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, United States
| | - Jichuan Wu
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, United States
| | - Vera P Krymskaya
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Jacob W Myerson
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, United States
| | - G Scott Worthen
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Jacob S Brenner
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
48
|
Wang Y, Carion TW, Ebrahim AS, Sosne G, Berger EA. Adjunctive Thymosin Beta-4 Treatment Influences PMN Effector Cell Function during Pseudomonas aeruginosa-Induced Corneal Infection. Cells 2021; 10:3579. [PMID: 34944086 PMCID: PMC8700181 DOI: 10.3390/cells10123579] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/14/2021] [Accepted: 12/15/2021] [Indexed: 11/18/2022] Open
Abstract
Previous work examining the therapeutic efficacy of adjunct thymosin beta 4 (Tβ4) to ciprofloxacin for ocular infectious disease has revealed markedly reduced inflammation (inflammatory mediators and innate immune cells) with increased activation of wound healing pathways. Understanding the therapeutic mechanisms of action have further revealed a synergistic effect with ciprofloxacin to enhance bacterial killing along with a regulatory influence over macrophage effector cell function. As a natural extension of the aforementioned work, the current study uses an experimental model of P. aeruginosa-induced keratitis to examine the influence of Tβ4 regarding polymorphonuclear leukocyte (PMN/neutrophil) cellular function, contributing to improved disease response. Flow cytometry was utilized to phenotypically profile infiltrating PMNs after infection. The generation of reactive oxygen species (ROS), neutrophil extracellular traps (NETs), and PMN apoptosis were investigated to assess the functional activities of PMNs in response to Tβ4 therapy. In vitro work using peritoneal-derived PMNs was similarly carried out to verify and extend our in vivo findings. The results indicate that the numbers of infiltrated PMNs into infected corneas were significantly reduced with adjunctive Tβ4 treatment. This was paired with the downregulated expression of proinflammatory markers on these cells, as well. Data generated from PMN functional studies suggested that the corneas of adjunctive Tβ4 treated B6 mice exhibit a well-regulated production of ROS, NETs, and limited PMN apoptosis. In addition to confirming the in vivo results, the in vitro findings also demonstrated that neutrophil elastase (NE) was unnecessary for NETosis. Collectively, these data provide additional evidence that adjunctive Tβ4 + ciprofloxacin treatment is a promising option for bacterial keratitis that addresses both the infectious pathogen and cellular-mediated immune response, as revealed by the current study.
Collapse
Affiliation(s)
- Yuxin Wang
- Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Thomas W Carion
- Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Abdul Shukkur Ebrahim
- Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Gabriel Sosne
- Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Elizabeth A Berger
- Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
49
|
Human Renal Fibroblasts, but Not Renal Epithelial Cells, Induce IL-1β Release during a Uropathogenic Escherichia coli Infection In Vitro. Cells 2021; 10:cells10123522. [PMID: 34944029 PMCID: PMC8700040 DOI: 10.3390/cells10123522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/07/2021] [Accepted: 12/11/2021] [Indexed: 11/17/2022] Open
Abstract
Understanding how uropathogenic Escherichia coli (UPEC) modulates the immune response in the kidney is essential to prevent UPEC from reaching the bloodstream and causing urosepsis. The purpose of this study was to elucidate if renal fibroblasts can release IL-1β during a UPEC infection and to investigate the mechanism behind the IL-1β release. We found that the UPEC strain CFT073 induced an increased IL-1β and LDH release from renal fibroblasts, but not from renal epithelial cells. The UPEC-induced IL-1β release was found to be NLRP3, caspase-1, caspase-4, ERK 1/2, cathepsin B and serine protease dependent in renal fibroblasts. We also found that the UPEC virulence factor α-hemolysin was necessary for IL-1β release. Conditioned medium from caspase-1, caspase-4 and NLRP3-deficient renal fibroblasts mediated an increased reactive oxygen species production from neutrophils, but reduced UPEC phagocytosis. Taken together, our study demonstrates that renal fibroblasts, but not renal epithelial cells, release IL-1β during a UPEC infection. This suggest that renal fibroblasts are vital immunoreactive cells and not only structural cells that produce and regulate the extracellular matrix.
Collapse
|
50
|
Ajendra J. Lessons in type 2 immunity: Neutrophils in Helminth infections. Semin Immunol 2021; 53:101531. [PMID: 34836773 DOI: 10.1016/j.smim.2021.101531] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 12/20/2022]
Abstract
Neutrophils constitute the body's first line of defense against invading pathogens. Equipped with a large array of tools, these immune cells are highly efficient in eliminating bacterial and viral infections, yet their activity can at the same time be detrimental to the host itself - this is the broad consensus on these granulocytes. However, the last decade has proven that neutrophils are a much more sophisticated cell type with unexpected and underappreciated functions in health and disease. In this review, we look at the latest discoveries in neutrophil biology with a focus on their role during the hallmark setting of type 2 immunity - helminth infection. We discuss the involvement of neutrophils in various helminth infection models and summarize the latest findings regarding neutrophil regulation and effector function. We will show that neutrophils have much more to offer than previously thought and while studies of neutrophils in helminth infections are still in its infancy, recent discoveries highlight more than ever that these cells are a key cog of the immune system, even during type 2 responses.
Collapse
Affiliation(s)
- Jesuthas Ajendra
- Lydia Becker Institute for Immunology & Infection, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK; Wellcome Centre for Cell-Matrix Research, Manchester, M13 9PT, UK.
| |
Collapse
|