1
|
Cardona-Ospina JA, Roy V, Marcano-Jiménez DE, Bos S, Duarte E, Zambrana JV, Bal A, Dias AG, Zhiteneva J, Huffaker J, Montenegro C, Kuan G, Ramos-Benitez MJ, Balmaseda A, Alter G, Harris E. IgA-driven neutrophil activation underlies post-Zika severe dengue disease in humans. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.11.25322002. [PMID: 40162272 PMCID: PMC11952487 DOI: 10.1101/2025.02.11.25322002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The four dengue virus serotypes (DENV1-4) and the related Zika flavivirus (ZIKV) are major public health concerns worldwide. Primary immunity against ZIKV increases the risk of a subsequent severe DENV2 infection, presenting a significant challenge for developing safe and effective ZIKV vaccines. However, the mechanisms driving this phenomenon remain unclear. Leveraging our long-standing Pediatric Dengue Cohort Study in Nicaragua, we show that serum anti-NS1 IgA antibodies elicited after a primary ZIKV infection drive neutrophil activation and correlate with increased risk of subsequent severe DENV2 disease. Depletion experiments combined with ex vivo functional NETosis assays confirmed that anti-NS1 IgA antibodies drive neutrophil activation in dengue hemorrhagic fever/dengue shock syndrome (DHF/DSS). Moreover, increased neutrophil degranulation in paired serum samples obtained during the acute DENV2 infection from the same individuals correlated with IgA binding to DENV2 NS1 and preceded the development of vascular leakage. This finding was corroborated in an orthogonal hospital-based study. Thus, serum anti-NS1 IgA enhances neutrophil activation in severe dengue, with implications for prognostics, therapeutics, and vaccines.
Collapse
Affiliation(s)
- Jaime A. Cardona-Ospina
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA
- Grupo Biomedicina, Facultad de Medicina, Institución Universitaria Visión de las Américas, Pereira, Colombia
| | - Vicky Roy
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA
| | - Dorca E. Marcano-Jiménez
- Department of Basic Sciences, Ponce Health Sciences University and Ponce Research Institute, Ponce, Puerto Rico
| | - Sandra Bos
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA
| | - Elias Duarte
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA
| | - José V. Zambrana
- Sustainable Sciences Institute, Managua, Nicaragua
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI
| | - Agamjot Bal
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA
| | - Antonio Gregorio Dias
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA
| | | | - Julia Huffaker
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA
| | | | - Guillermina Kuan
- Sustainable Sciences Institute, Managua, Nicaragua
- Centro de Salud Sócrates Flores Vivas, Ministerio de Salud, Managua, Nicaragua
| | - Marcos J. Ramos-Benitez
- Department of Basic Sciences, Ponce Health Sciences University and Ponce Research Institute, Ponce, Puerto Rico
| | - Angel Balmaseda
- Sustainable Sciences Institute, Managua, Nicaragua
- Laboratorio Nacional de Virología, Centro Nacional de Diagnóstico y Referencia, Ministerio de Salud, Managua, Nicaragua
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA
| |
Collapse
|
2
|
Abo-Zaid OAR, Moawed FSM, Eldin ES, Farrag MA, Ahmed ESA. Antitumor activity of gamma-irradiated Rosa canina L. against lung carcinoma in rat model: a proposed mechanism. BMC Complement Med Ther 2025; 25:86. [PMID: 40022036 PMCID: PMC11869437 DOI: 10.1186/s12906-025-04813-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 02/05/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Lung cancer is one of the most prevalent malignancies globally and is the leading cause of cancer-related mortality. Although cisplatin is a widely utilized chemotherapeutic agent, its clinical efficacy is often hampered by significant toxicity and undesirable side effects. Rosa canina, a medicinal plant, has demonstrated a range of beneficial biological activities, including anti-inflammatory, anticancer, immunomodulatory, antioxidant, and genoprotective effects. METHODS This study aimed to investigate the potential of Rosa canina to enhance the anticancer efficacy of cisplatin in a dimethyl benz(a)anthracene-induced lung cancer model using female rats. The animals were administered Rosa canina, cisplatin, or a combination of both treatments. The expression levels of critical signaling molecules were evaluated, including phosphoinositide-3-kinase (PI3K), Akt, mammalian target of rapamycin (mTOR), cleaved poly (ADP-ribose) polymerase (PARP-1), myeloid differentiation factor 88 (MyD88), and tumor necrosis factor receptor-associated factor (TRAF), in addition to various autophagic markers. Furthermore, we assessed the levels of toll-like receptor 2 (TLR2), nuclear factor kappa B (NF-κB), and apoptotic markers in lung tissue, complemented by histopathological examinations. RESULTS The combined treatment of Rosa canina extract and cisplatin significantly inhibited lung cancer cell proliferation by downregulating PARP-1 and the TLR2/MyD88/TRAF6/NF-κB signaling pathway, as well as the PI3K/Akt/mTOR pathway. Moreover, this combination therapy promoted autophagy and apoptosis, evidenced by elevated levels of autophagic and apoptotic markers. CONCLUSION Overall, the findings of this study suggest that Rosa canina enhances the anticancer effects of cisplatin by inhibiting cancer cell proliferation while simultaneously inducing autophagy and apoptosis. Thus, Rosa can be used as adjuvant to cisplatin chemotherapy to overcome its limitations which may be considered a new approach during lung cancer treatment strategy.
Collapse
Affiliation(s)
- Omayma A R Abo-Zaid
- Biochemistry and Molecular Biology Department, Faculty of Vet. Med, Benha University, Benha, Egypt
| | - Fatma S M Moawed
- Health Radiation Research, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Eman S Eldin
- Health Radiation Research, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Mostafa A Farrag
- Radiation Biology , National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Nasr City, Cairo, 11787, Egypt
| | - Esraa S A Ahmed
- Radiation Biology , National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Nasr City, Cairo, 11787, Egypt.
| |
Collapse
|
3
|
Kim YH, Ahn HJ, Kim HS, Nam HW. Small molecule kinase inhibitor altiratinib inhibits brain cyst forming bradyzoites of Toxoplasma gondii. J Microbiol 2025; 63:e2409001. [PMID: 40044130 DOI: 10.71150/jm.2409001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 01/10/2025] [Indexed: 05/13/2025]
Abstract
Chronic toxoplasmosis is caused by Toxoplasma gondii bradyzoites. This study assessed six candidate small molecule kinase inhibitors (SMKIs) against bradyzoites (ME49 strain), the reactivated form of the parasite resulting from the rupture of brain cysts. Bradyzoites were obtained from mouse brain cysts, cultured in ARPE-19 cells, and treated with afatinib and neratinib (HER2/HER4 inhibitors), ACTB-1003 and regorafenib (VEGFR-2 inhibitors), or altiratinib and foretinib (c-MET inhibitors). The effects on the growth of T. gondii were analyzed by western blot and immunofluorescence assay. Changes in the host cells were assessed using markers for cell viability, apoptosis, necrosis, and autophagy. All inhibitors blocked the growth of bradyzoites, although afatinib was less effective. Afatinib enhanced autophagy signals, while ACTB-1003 and neratinib affected mitochondrial biosynthesis and mitophagy. Altiratinib demonstrated an effect against bradyzoites at the lowest concentration with minimal impact on the host cells. It may be effective in blocking the reactivation of brain cysts in immunodeficiency patients caused by bradyzoites.
Collapse
Affiliation(s)
- Yeong Hoon Kim
- Department of Ophthalmology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Hye-Jin Ahn
- Department of Parasitology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Hwa Sun Kim
- Department of Family Medicine, Veterans Health Service Medical Center, Seoul 05368, Republic of Korea
| | - Ho-Woo Nam
- Department of Parasitology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
4
|
Goris M, Passelli K, Peyvandi S, Díaz-Varela M, Billion O, Prat-Luri B, Demarco B, Desponds C, Termote M, Iniguez E, Dey S, Malissen B, Kamhawi S, Hurrell BP, Broz P, Tacchini-Cottier F. NLRP1-dependent activation of Gasdermin D in neutrophils controls cutaneous leishmaniasis. PLoS Pathog 2024; 20:e1012527. [PMID: 39250503 PMCID: PMC11412672 DOI: 10.1371/journal.ppat.1012527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 09/19/2024] [Accepted: 08/22/2024] [Indexed: 09/11/2024] Open
Abstract
Intracellular pathogens that replicate in host myeloid cells have devised ways to inhibit the cell's killing machinery. Pyroptosis is one of the host strategies used to reduce the pathogen replicating niche and thereby control its expansion. The intracellular Leishmania parasites can survive and use neutrophils as a silent entry niche, favoring subsequent parasite dissemination into the host. Here, we show that Leishmania mexicana induces NLRP1- and caspase-1-dependent Gasdermin D (GSDMD)-mediated pyroptosis in neutrophils, a process critical to control the parasite-induced pathology. In the absence of GSDMD, we observe an increased number of infected dermal neutrophils two days post-infection. Using adoptive neutrophil transfer in neutropenic mice, we show that pyroptosis contributes to the regulation of the neutrophil niche early after infection. The critical role of neutrophil pyroptosis and its positive influence on the regulation of the disease outcome was further demonstrated following infection of mice with neutrophil-specific deletion of GSDMD. Thus, our study establishes neutrophil pyroptosis as a critical regulator of leishmaniasis pathology.
Collapse
Affiliation(s)
- Michiel Goris
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| | - Katiuska Passelli
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| | - Sanam Peyvandi
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| | - Miriam Díaz-Varela
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| | - Oaklyne Billion
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| | - Borja Prat-Luri
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| | - Benjamin Demarco
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Chantal Desponds
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| | - Manon Termote
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| | - Eva Iniguez
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Somaditya Dey
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
- Post Graduate Department of Zoology, Barasat Government College, Barasat, West Bengal, India
| | - Bernard Malissen
- INSERM, CNRS, Centre D’Immunologie de Marseille-Luminy, Aix-Marseille Université, Marseille, France
| | - Shaden Kamhawi
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Benjamin P. Hurrell
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Petr Broz
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Fabienne Tacchini-Cottier
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
5
|
Barbosa-Matos C, Borges-Pereira C, Libório-Ramos S, Fernandes R, Oliveira M, Mendes-Frias A, Silvestre R, Osório NS, Bastos HN, Santos RF, Guimarães S, Morais A, Mazzone M, Carvalho A, Cunha C, Costa S. Deregulated immune cell recruitment orchestrated by c-MET impairs pulmonary inflammation and fibrosis. Respir Res 2024; 25:257. [PMID: 38909206 PMCID: PMC11193258 DOI: 10.1186/s12931-024-02884-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 06/13/2024] [Indexed: 06/24/2024] Open
Abstract
BACKGROUND Pulmonary fibrosis (PF) represents the pathologic end stage of several interstitial lung diseases (ILDs) associated with high morbidity and mortality rates. However, current treatments can only delay disease progression rather than provide a cure. The role of inflammation in PF progression is well-established, but new insights into immune regulation are fundamental for developing more efficient therapies. c-MET signaling has been implicated in the migratory capacity and effector functions of immune cells. Nevertheless, the role of this signaling pathway in the context of PF-associated lung diseases remains unexplored. METHODS To determine the influence of c-MET in immune cells in the progression of pulmonary fibrosis, we used a conditional deletion of c-Met in immune cells. To induce pulmonary fibrosis mice were administered with bleomycin (BLM) intratracheally. Over the course of 21 days, mice were assessed for weight change, and after euthanasia at different timepoints, bronchoalveolar lavage fluid cells and lung tissue were assessed for inflammation and fibrosis. Furthermore, c-MET expression was assessed in cryobiopsy sections, bronchoalveolar lavage fluid cells samples and single cell RNA-sequencing dataset from human patients with distinct interstitial lung diseases. RESULTS c-MET expression was induced in lung immune cells, specifically in T cells, interstitial macrophages, and neutrophils, during the inflammatory phase of BLM-induced PF mouse model. Deletion of c-Met in immune cells correlated with earlier weight recovery and improved survival of BLM-treated mice. Moreover, the deletion of c-Met in immune cells was associated with early recruitment of the immune cell populations, normally found to express c-MET, leading to a subsequent attenuation of the cytotoxic and proinflammatory environment. Consequently, the less extensive inflammatory response, possibly coupled with tissue repair, culminated in less exacerbated fibrotic lesions. Furthermore, c-MET expression was up-regulated in lung T cells from patients with fibrosing ILD, suggesting a potential involvement of c-MET in the development of fibrosing disease. CONCLUSIONS These results highlight the critical contribution of c-MET signaling in immune cells to their enhanced uncontrolled recruitment and activation toward a proinflammatory and profibrotic phenotype, leading to the exacerbation of lung injury and consequent development of fibrosis.
Collapse
Affiliation(s)
- Catarina Barbosa-Matos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, Braga, 4710-057, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Caroline Borges-Pereira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, Braga, 4710-057, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Sofia Libório-Ramos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, Braga, 4710-057, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Raquel Fernandes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, Braga, 4710-057, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Marcela Oliveira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, Braga, 4710-057, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ana Mendes-Frias
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, Braga, 4710-057, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ricardo Silvestre
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, Braga, 4710-057, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Nuno S Osório
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, Braga, 4710-057, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Hélder N Bastos
- Department of Pneumology, Centro Hospitalar do São João, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
| | - Rita F Santos
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- School of Health Sciences - Polytechnic of Porto, Porto, Portugal
| | - Susana Guimarães
- Department of Pathology, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - António Morais
- Department of Pneumology, Centro Hospitalar do São João, Porto, Portugal
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Louvain, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, KU Leuven, Louvain, Belgium
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, Braga, 4710-057, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Cristina Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, Braga, 4710-057, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Sandra Costa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, Braga, 4710-057, Portugal.
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
6
|
Díaz-Varela M, Sanchez-Hidalgo A, Calderon-Copete S, Tacchini V, Shipley TR, Ramírez LG, Marquis J, Fernández OL, Saravia NG, Tacchini-Cottier F. The different impact of drug-resistant Leishmania on the transcription programs activated in neutrophils. iScience 2024; 27:109773. [PMID: 38711445 PMCID: PMC11070714 DOI: 10.1016/j.isci.2024.109773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/22/2024] [Accepted: 04/15/2024] [Indexed: 05/08/2024] Open
Abstract
Drug resistance threatens the effective control of infections, including parasitic diseases such as leishmaniases. Neutrophils are essential players in antimicrobial control, but their role in drug-resistant infections is poorly understood. Here, we evaluated human neutrophil response to clinical parasite strains having distinct natural drug susceptibility. We found that Leishmania antimony drug resistance significantly altered the expression of neutrophil genes, some of them transcribed by specific neutrophil subsets. Infection with drug-resistant parasites increased the expression of detoxification pathways and reduced the production of cytokines. Among these, the chemokine CCL3 was predominantly impacted, which resulted in an impaired ability of neutrophils to attract myeloid cells. Moreover, decreased myeloid recruitment when CCL3 levels are reduced was confirmed by blocking CCL3 in a mouse model. Collectively, these findings reveal that the interplay between naturally drug-resistant parasites and neutrophils modulates the infected skin immune microenvironment, revealing a key role of neutrophils in drug resistance.
Collapse
Affiliation(s)
- Míriam Díaz-Varela
- Department of Immunobiology, WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, 1066 Epalinges, Switzerland
| | - Andrea Sanchez-Hidalgo
- Centro Internacional de Entrenamiento e Investigaciones Médicas, CIDEIM, Cali 760031, Colombia
- Universidad Icesi, Cali 760031, Colombia
| | - Sandra Calderon-Copete
- Lausanne Genomic Technologies Facility, University of Lausanne, 1015 Lausanne, Switzerland
| | - Virginie Tacchini
- Department of Immunobiology, WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, 1066 Epalinges, Switzerland
| | - Tobias R. Shipley
- Department of Immunobiology, WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, 1066 Epalinges, Switzerland
| | - Lady Giovanna Ramírez
- Centro Internacional de Entrenamiento e Investigaciones Médicas, CIDEIM, Cali 760031, Colombia
- Universidad Icesi, Cali 760031, Colombia
| | - Julien Marquis
- Lausanne Genomic Technologies Facility, University of Lausanne, 1015 Lausanne, Switzerland
| | - Olga Lucía Fernández
- Centro Internacional de Entrenamiento e Investigaciones Médicas, CIDEIM, Cali 760031, Colombia
- Universidad Icesi, Cali 760031, Colombia
| | - Nancy Gore Saravia
- Centro Internacional de Entrenamiento e Investigaciones Médicas, CIDEIM, Cali 760031, Colombia
- Universidad Icesi, Cali 760031, Colombia
| | - Fabienne Tacchini-Cottier
- Department of Immunobiology, WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, 1066 Epalinges, Switzerland
| |
Collapse
|
7
|
Jing W, Wang G, Cui Z, Li X, Zeng S, Jiang X, Li W, Han B, Xing N, Zhao Y, Chen S, Shi B. Tumor-neutrophil cross talk orchestrates the tumor microenvironment to determine the bladder cancer progression. Proc Natl Acad Sci U S A 2024; 121:e2312855121. [PMID: 38713626 PMCID: PMC11098120 DOI: 10.1073/pnas.2312855121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 03/13/2024] [Indexed: 05/09/2024] Open
Abstract
The immune landscape of bladder cancer progression is not fully understood, and effective therapies are lacking in advanced bladder cancer. Here, we visualized that bladder cancer cells recruited neutrophils by secreting interleukin-8 (IL-8); in turn, neutrophils played dual functions in bladder cancer, including hepatocyte growth factor (HGF) release and CCL3highPD-L1high super-immunosuppressive subset formation. Mechanistically, c-Fos was identified as the mediator of HGF up-regulating IL-8 transcription in bladder cancer cells, which was central to the positive feedback of neutrophil recruitment. Clinically, compared with serum IL-8, urine IL-8 was a better biomarker for bladder cancer prognosis and clinical benefit of immune checkpoint blockade (ICB). Additionally, targeting neutrophils or hepatocyte growth factor receptor (MET) signaling combined with ICB inhibited bladder cancer progression and boosted the antitumor effect of CD8+ T cells in mice. These findings reveal the mechanism by which tumor-neutrophil cross talk orchestrates the bladder cancer microenvironment and provide combination strategies, which may have broad impacts on patients suffering from malignancies enriched with neutrophils.
Collapse
Affiliation(s)
- Weiqiang Jing
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province250012, China
| | - Ganyu Wang
- Department of Pediatric Surgery, Qilu Hospital Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province250012, China
| | - Zhiwei Cui
- Department of Immunology, Shandong Provincial Key Laboratory of Infection & Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province250012, China
| | - Xinyuan Li
- Department of Immunology, Shandong Provincial Key Laboratory of Infection & Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province250012, China
| | - Shuyan Zeng
- Department of Immunology, Shandong Provincial Key Laboratory of Infection & Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province250012, China
| | - Xin Jiang
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province250012, China
| | - Wushan Li
- Department of Obstetrics, Jinan Maternity and Child Care Hospital Shandong First Medical University, Jinan, Shandong Province250000, China
| | - Bo Han
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province250012, China
| | - Nianzeng Xing
- Department of Urology and State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing10021, China
| | - Yunxue Zhao
- Department of Immunology, Shandong Provincial Key Laboratory of Infection & Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province250012, China
| | - Shouzhen Chen
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province250012, China
| | - Benkang Shi
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province250012, China
| |
Collapse
|
8
|
Fernández OL, Rosales-Chilama M, Sánchez-Hidalgo A, Gómez P, Rebellón-Sánchez DE, Regli IB, Díaz-Varela M, Tacchini-Cottier F, Saravia NG. Natural resistance to meglumine antimoniate is associated with treatment failure in cutaneous leishmaniasis caused by Leishmania (Viannia) panamensis. PLoS Negl Trop Dis 2024; 18:e0012156. [PMID: 38709850 PMCID: PMC11098511 DOI: 10.1371/journal.pntd.0012156] [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: 12/19/2023] [Revised: 05/16/2024] [Accepted: 04/17/2024] [Indexed: 05/08/2024] Open
Abstract
The multifactorial basis of therapeutic response can obscure the relation between antimicrobial drug susceptibility and clinical outcome. To discern the relationship between parasite susceptibility to meglumine antimoniate (SbV) and therapeutic outcome of cutaneous leishmaniasis, risk factors for treatment failure were considered in evaluating this relationship in ninety-one cutaneous leishmaniasis patients and corresponding clinical strains of Leishmania (Viannia) panamensis. Parasite susceptibility to 32 μg SbV/mL (plasma Cmax) was evaluated in primary human macrophages, PBMCs, and U937 macrophages. Early parasitological response to treatment was determined in lesions of a subgroup of patients, and pathogenicity of Sb-resistant and sensitive clinical strains was compared in BALB/c mice. Parasite survival in cell models and patient lesions was determined by qRT-PCR of Leishmania 7SLRNA transcript. Parasite loads in BALB/c mice were quantified by limiting dilution analysis. The disparate Sb-susceptibility of parasite subpopulations distinguished by isoenzyme profiles (zymodemes) was manifest in all cell models. Notably, Sb-resistance defined by parasite survival, was most effectively discerned in U937 macrophages compared with primary human host cells, significantly higher among strains from patients who failed treatment than cured and, significantly associated with treatment failure. Each unit increase in transformed survival rate corresponded to a 10.6-fold rise in the odds of treatment failure. Furthermore, treatment failure was significantly associated with naturally Sb-resistant zymodeme 2.3 strains, which also produced larger lesions and parasite burdens in BALB/c mice than Sb-sensitive zymodeme 2.2 strains. The confounding effect of host risk factors for treatment failure in discerning this association was evidenced in comparing strains from patients with and without the defined risk factors for treatment failure. These results establish the association of natural resistance to meglumine antimoniate with treatment failure, the importance of host risk factors in evaluating drug susceptibility and treatment outcome, and the clinical and epidemiological relevance of natural Sb-resistance in L. (V.) panamensis subpopulations.
Collapse
Affiliation(s)
- Olga Lucía Fernández
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
- Universidad Icesi, Cali, Colombia
| | - Mariana Rosales-Chilama
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
- Universidad Icesi, Cali, Colombia
| | - Andrea Sánchez-Hidalgo
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
- Universidad Icesi, Cali, Colombia
| | - Paola Gómez
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
- Universidad Icesi, Cali, Colombia
| | - David Esteban Rebellón-Sánchez
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
- Fundación Valle del Lili, Centro de Investigaciones Clínicas (CIC), Cali, Colombia
| | - Ivo B. Regli
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Míriam Díaz-Varela
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | | | - Nancy Gore Saravia
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
- Universidad Icesi, Cali, Colombia
| |
Collapse
|
9
|
Wang S, Wang L, Qiu M, Lin Z, Qi W, Lv J, Wang Y, Lu Y, Li X, Chen W, Qiu W. Constructing and validating a risk model based on neutrophil-related genes for evaluating prognosis and guiding immunotherapy in colon cancer. J Gene Med 2024; 26:e3684. [PMID: 38618694 DOI: 10.1002/jgm.3684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/25/2024] [Accepted: 03/03/2024] [Indexed: 04/16/2024] Open
Abstract
BACKGROUND Colon cancer is one of the most common digestive tract malignancies. Although immunotherapy has brought new hope to colon cancer patients, there is still a large proportion of patients who do not benefit from immunotherapy. Studies have shown that neutrophils can interact with immune cells and immune factors to affect the prognosis of patients. METHODS We first determined the infiltration level of neutrophils in tumors using the CIBERSORT algorithm and identified key genes in the final risk model by Spearman correlation analysis and subsequent Cox analysis. The risk score of each patient was obtained by multiplying the Cox regression coefficient and the gene expression level, and patients were divided into two groups based on the median of risk score. Differences in overall survival (OS) and progression-free survival (PFS) were assessed by Kaplan-Meier survival analysis, and model accuracy was validated in independent dataset. Differences in immune infiltration and immunotherapy were evaluated by immunoassay. Finally, immunohistochemistry and western blotting were performed to verify the expression of the three genes in the colon normal and tumor tissues. RESULTS We established and validated a risk scoring model based on neutrophil-related genes in two independent datasets, The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) database, with SLC11A1 and SLC2A3 as risk factors and MMP3 as a protective factor. A new nomogram was constructed and validated by combining clinical characteristics and the risk score model to better predict patients OS and PFS. Immune analysis showed that patients in the high-risk group had immune cell infiltration level, immune checkpoint level and tumor mutational burden, and were more likely to benefit from immunotherapy. CONCLUSIONS The low-risk group showed better OS and PFS than the high-risk group in the neutrophil-related gene-based risk model. Patients in the high-risk group presented higher immune infiltration levels and tumor mutational burden and thus may be more responsive to immunotherapy.
Collapse
Affiliation(s)
- Shasha Wang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lili Wang
- Department of Oncology, Rizhao Central Hospital, Rizhao, China
| | - Mingxiu Qiu
- Department Second of Respiratory and Critical Care, Qingdao Municipal Hospital, Qingdao, China
| | - Zhongkun Lin
- Department of Oncology, Shandong Provincial Third Hospital, Shandong University, Jinan, China
| | - Weiwei Qi
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jing Lv
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yan Wang
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yangyang Lu
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaoxuan Li
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wenzhi Chen
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Wensheng Qiu
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|