1
|
Alves IA, Staudt KJ, Torres BGS, Oliveira CE, Rieper RLA, Kowalski L, de Araújo BV. Does Cryptococcus neoformans infection alter antifungal distribution: an animal model exploring pharmacokinetic changes. Future Microbiol 2025; 20:489-498. [PMID: 40178503 PMCID: PMC11980448 DOI: 10.1080/17460913.2025.2484956] [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: 12/04/2024] [Accepted: 03/24/2025] [Indexed: 04/05/2025] Open
Abstract
AIM Assessing the disseminated meningitis caused by Cryptoccocus neoformans in Wistar rats and its impact on antifungal distribution by microdialysis (µD). MATERIALS & METHODS The yeast presence was investigated in different tissues by histological and microbiological assays, and biochemical parameters such as urea, glutamate oxaloacetate transaminase (GOT), glutamate pyruvate transaminase (GPT), creatinine, creatine kinase (CK), creatinine, albumin level, leukocyte counts, and brain vascular permeability (Evans blue test) were evaluated in healthy and infected groups. Levels of fluconazole reached in the animal's brain were determined by µD. RESULTS Differences in albumin, urea, GPT, and CK between healthy and infected animals were observed in the levels of Evans blue as well as in the brain (0.51 vs 1.50 µg/gbrain). The drugs' distribution in the brain of infected animals was higher than that in the brain of healthy ones (ft = 1.37 vs ft = 0.54). CONCLUSION The model validated presents characteristics similar to those observed in patients and can be applied to pharmacokinetic investigations.
Collapse
Affiliation(s)
- Izabel Almeida Alves
- Faculdade de Farmácia, Departamento do Medicamento, Universidade Federal da Bahia, Salvador, Bahia, Brasil
- Programa de Pós-Graduação em Farmácia, Universidade Estadual da Bahia, Salvador, Bahia, Brasil
- Programa de Pós-graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brasil
| | - Keli Jaqueline Staudt
- Programa de Pós-graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brasil
- Departamento de Ciências da Saúde, Universidade Regional Integrada do Alto Uruguai e das Missões, Santo Ângelo, Rio Grande do Sul, Brasil
| | - Bruna Gaelzer Silva Torres
- Programa de Pós-graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brasil
| | | | - Ryan Lago Araujo Rieper
- Faculdade de Farmácia, Departamento do Medicamento, Universidade Federal da Bahia, Salvador, Bahia, Brasil
| | - Layza Kowalski
- Programa de Pós-graduação em Ciências Biológicas: Fisiologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brasil
| | - Bibiana Verlindo de Araújo
- Programa de Pós-graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brasil
| |
Collapse
|
2
|
Munzen ME, Mathew C, Enriquez V, Minhas A, Charles-Niño CL, Saytoo D, Reguera-Gomez M, Dores MR, Martinez LR. Inhibition of RhoA Prevents Cryptococcus neoformans Capsule Glucuronoxylomannan-Stimulated Brain Endothelial Barrier Disruption. J Infect Dis 2024; 230:1042-1051. [PMID: 38622836 PMCID: PMC11481333 DOI: 10.1093/infdis/jiae187] [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: 12/13/2023] [Revised: 04/03/2024] [Accepted: 04/10/2024] [Indexed: 04/17/2024] Open
Abstract
Cryptococcus neoformans (Cn) is an opportunistic fungus that causes severe central nervous system (CNS) disease in immunocompromised individuals. Brain parenchyma invasion requires fungal traversal of the blood-brain barrier. In this study, we describe that Cn alters the brain endothelium by activating small GTPase RhoA, causing reorganization of the actin cytoskeleton and tight junction modulation to regulate endothelial barrier permeability. We confirm that the main fungal capsule polysaccharide glucuronoxylomannan is responsible for these alterations. We reveal a therapeutic benefit of RhoA inhibition by CCG-1423 in vivo. RhoA inhibition prolonged survival and reduced fungal burden in a murine model of disseminated cryptococcosis, supporting the therapeutic potential of targeting RhoA in the context of cryptococcal infection. We examine the complex virulence of Cn in establishing CNS disease, describing cellular components of the brain endothelium that may serve as molecular targets for future antifungal therapies to alleviate the burden of life-threatening cryptococcal CNS infection.
Collapse
Affiliation(s)
- Melissa E Munzen
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville
| | - Cristian Mathew
- Department of Biology, Hofstra University, Hempstead, New York
| | - Vanessa Enriquez
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville
| | - Amanjeet Minhas
- Department of Biology, Hofstra University, Hempstead, New York
| | | | | | - Marta Reguera-Gomez
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville
| | - Michael R Dores
- Department of Biology, Hofstra University, Hempstead, New York
| | - Luis R Martinez
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville
- Emerging Pathogens Institute
- Center for Immunology and Transplantation
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville
| |
Collapse
|
3
|
Dong L, Zhuang X, Yang T, Yan K, Cai Y. A physiologically based pharmacokinetic model of voriconazole in human CNS-Integrating time-dependent inhibition of CYP3A4, genetic polymorphisms of CYP2C19 and possible transporter mechanisms. Int J Antimicrob Agents 2024; 64:107310. [PMID: 39168418 DOI: 10.1016/j.ijantimicag.2024.107310] [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: 03/04/2024] [Revised: 07/26/2024] [Accepted: 08/12/2024] [Indexed: 08/23/2024]
Abstract
OBJECTIVES Voriconazole is a classical antifungal drug that is often used to treat CNS fungal infections due to its permeability through the BBB. However, its clinical use remains challenging because of its narrow therapeutic window and wide inter-individual variability. In this study, we proposed an optimised and validated PBPK model by integrating in vitro, in vivo and clinical data to simulate the distribution and PK process of voriconazole in the CNS, providing guidance for clinical individualised treatment. METHODS The model structure was optimised and tissue-to-plasma partition coefficients were obtained through animal experiments. Using the allometric relationships, the distribution of voriconazole in the human CNS was predicted. The model integrated factors affecting inter-individual variation and drug interactions of voriconazole-polymorphisms in the CYP2C19 gene and auto-inhibition and then was validated using real clinical data. RESULTS The overall AFE value showing model predicted differences was 1.1420 in the healthy population; and in the first prediction of plasma and CSF in actual clinical patients, 89.5% of the values were within the 2-fold error interval, indicating good predictive performance of the model. The bioavailability of voriconazole varied at different doses (39%-86%), and the optimised model conformed to this pattern (46%-83%). CONCLUSIONS Combined with the relevant pharmacodynamic indexes, the PBPK model provides a feasible way for precise medication in patients with CNS infection and improve the treatment effect and prognosis.
Collapse
Affiliation(s)
- Liuhan Dong
- Center of Medicine Clinical Research, Department of Pharmacy, Chinese PLA General Hospital, Beijing, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Xiaomei Zhuang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Tianli Yang
- Center of Medicine Clinical Research, Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
| | - Kaicheng Yan
- Center of Medicine Clinical Research, Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
| | - Yun Cai
- Center of Medicine Clinical Research, Department of Pharmacy, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
4
|
Soraci L, Beccacece A, Princiotto M, Villalta Savedra E, Gambuzza ME, Aguennouz M, Corsonello A, Luciani F, Muglia L, Filicetti E, Greco GI, Volpentesta M, Biscetti L. The emerging links between immunosenescence in innate immune system and neurocryptococcosis. Front Immunol 2024; 15:1410090. [PMID: 39229268 PMCID: PMC11369721 DOI: 10.3389/fimmu.2024.1410090] [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: 03/31/2024] [Accepted: 07/29/2024] [Indexed: 09/05/2024] Open
Abstract
Immunosenescence refers to the age-related progressive decline of immune function contributing to the increased susceptibility to infectious diseases in older people. Neurocryptococcosis, an infectious disease of central nervous system (CNS) caused by Cryptococcus neoformans (C. Neoformans) and C. gattii, has been observed with increased frequency in aged people, as result of the reactivation of a latent infection or community acquisition. These opportunistic microorganisms belonging to kingdom of fungi are capable of surviving and replicating within macrophages. Typically, cryptococcus is expelled by vomocytosis, a non-lytic expulsive mechanism also promoted by interferon (IFN)-I, or by cell lysis. However, whereas in a first phase cryptococcal vomocytosis leads to a latent asymptomatic infection confined to the lung, an enhancement in vomocytosis, promoted by IFN-I overproduction, can be deleterious, leading the fungus to reach the blood stream and invade the CNS. Cryptococcus may not be easy to diagnose in older individuals and, if not timely treated, could be potentially lethal. Therefore, this review aims to elucidate the putative causes of the increased incidence of cryptococcal CNS infection in older people discussing in depth the mechanisms of immunosenscence potentially able to predispose to neurocryptococcosis, laying the foundations for future research. A deepest understanding of this relationship could provide new ways to improve the prevention and recognition of neurocryptococcosis in aged frail people, in order to quickly manage pharmacological interventions and to adopt further preventive measures able to reduce the main risk factors.
Collapse
Affiliation(s)
- Luca Soraci
- Unit of Geriatric Medicine, Italian National Research Center on Aging (IRCCS INRCA), Cosenza, Italy
| | - Alessia Beccacece
- Centre for Biostatistics and Applied Geriatric Clinical Epidemiology, Italian National Research Center on Aging (IRCCS INRCA), Ancona, Italy
| | | | | | | | - M’Hammed Aguennouz
- Department of Clinical and Experimental Medicine, Unit of Neurology and Neuromuscular Diseases, University of Messina, Messina, Italy
| | - Andrea Corsonello
- Unit of Geriatric Medicine, Italian National Research Center on Aging (IRCCS INRCA), Cosenza, Italy
- Department of Pharmacy, Health and Nutritional Sciences, School of Medicine and Digital Technologies, University of Calabria, Arcavacata di Rende, Italy
| | | | - Lucia Muglia
- Centre for Biostatistics and Applied Geriatric Clinical Epidemiology, Italian National Research Center on Aging (IRCCS INRCA), Cosenza, Italy
| | - Elvira Filicetti
- Unit of Geriatric Medicine, Italian National Research Center on Aging (IRCCS INRCA), Cosenza, Italy
| | - Giada Ida Greco
- Unit of Geriatric Medicine, Italian National Research Center on Aging (IRCCS INRCA), Cosenza, Italy
| | - Mara Volpentesta
- Unit of Geriatric Medicine, Italian National Research Center on Aging (IRCCS INRCA), Cosenza, Italy
| | - Leonardo Biscetti
- Section of Neurology, Italian National Research Center on Aging (IRCCS INRCA), Ancona, Italy
| |
Collapse
|
5
|
Qin J, Nong L, Zhu Q, Huang Z, Wu F, Li S. A Retrospective Analysis of Central and Peripheral Metabolic Characteristics in Patients with Cryptococcal Meningitis. Neurol Ther 2024; 13:763-784. [PMID: 38643256 PMCID: PMC11136911 DOI: 10.1007/s40120-024-00610-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 03/19/2024] [Indexed: 04/22/2024] Open
Abstract
INTRODUCTION Most current treatment strategies and investigations on cryptococcal meningitis (CM) focus primarily on the central nervous system (CNS), often overlooking the complex interplay between the CNS and the peripheral system. This study aims to explore the characteristics of central and peripheral metabolism in patients with CM. METHODS Patients diagnosed with CM as per the hospital records of the Fourth People's Hospital of Nanning were retrospectively analyzed. Patients were divided into two groups, non-structural damage of the brain (NSDB) and structural damage of the brain (SDB), according to the presence of brain lesions as detected with imaging. Based on the presence of enlarged cerebral ventricles, the cases in the SDB group were classified into non-ventriculomegaly (NVM) and ventriculomegaly (VM). Various parameters of cerebrospinal fluid (CSF) and peripheral blood (PB) were analyzed. RESULTS A significant correlation was detected between CSF and PB parameters. The levels of CSF-adenosine dehydrogenase (ADA), CSF-protein, CSF-glucose, and CSF-chloride ions were significantly correlated with the levels of PB-aminotransferase, PB-bilirubin, PB-creatinine (Cr), PB-urea nitrogen, PB-electrolyte, PB-protein, and PB-lipid. Compared with NSDB, the levels of CSF-glucose were significantly decreased in the SDB group, while the levels of CSF-lactate dehydrogenase (LDH) and CSF-protein were significantly increased in the SDB group. In the SDB group, the levels of PB-potassium, PB-hemoglobin(Hb), and PB-albumin were significantly decreased in the patients with VM, while the level of PB-urea nitrogen was significantly increased in these patients. CONCLUSION Metabolic and structural alterations in the brain may be associated with peripheral metabolic changes.
Collapse
Affiliation(s)
- Jianglong Qin
- The Fourth People's Hospital of Nanning, 1 Chang-Gang-Two-Li Road, Nanning, Guangxi, 530023, People's Republic of China
| | - Lanwei Nong
- The Fourth People's Hospital of Nanning, 1 Chang-Gang-Two-Li Road, Nanning, Guangxi, 530023, People's Republic of China
| | - Qingdong Zhu
- The Fourth People's Hospital of Nanning, 1 Chang-Gang-Two-Li Road, Nanning, Guangxi, 530023, People's Republic of China
| | - Zhizhong Huang
- The Fourth People's Hospital of Nanning, 1 Chang-Gang-Two-Li Road, Nanning, Guangxi, 530023, People's Republic of China
| | - Fengyao Wu
- The Fourth People's Hospital of Nanning, 1 Chang-Gang-Two-Li Road, Nanning, Guangxi, 530023, People's Republic of China
| | - Sijun Li
- The Fourth People's Hospital of Nanning, 1 Chang-Gang-Two-Li Road, Nanning, Guangxi, 530023, People's Republic of China.
| |
Collapse
|
6
|
Gopinadhan A, Hughes JM, Conroy AL, John CC, Canfield SG, Datta D. A human pluripotent stem cell-derived in vitro model of the blood-brain barrier in cerebral malaria. Fluids Barriers CNS 2024; 21:38. [PMID: 38693577 PMCID: PMC11064301 DOI: 10.1186/s12987-024-00541-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 04/18/2024] [Indexed: 05/03/2024] Open
Abstract
BACKGROUND Blood-brain barrier (BBB) disruption is a central feature of cerebral malaria (CM), a severe complication of Plasmodium falciparum (Pf) infections. In CM, sequestration of Pf-infected red blood cells (Pf-iRBCs) to brain endothelial cells combined with inflammation, hemolysis, microvasculature obstruction and endothelial dysfunction mediates BBB disruption, resulting in severe neurologic symptoms including coma and seizures, potentially leading to death or long-term sequelae. In vitro models have advanced our knowledge of CM-mediated BBB disruption, but their physiological relevance remains uncertain. Using human induced pluripotent stem cell-derived brain microvascular endothelial cells (hiPSC-BMECs), we aimed to develop a novel in vitro model of the BBB in CM, exhibiting enhanced barrier properties. METHODS hiPSC-BMECs were co-cultured with HB3var03 strain Pf-iRBCs up to 9 h. Barrier integrity was measured using transendothelial electrical resistance (TEER) and sodium fluorescein permeability assays. Localization and expression of tight junction (TJ) proteins (occludin, zonula occludens-1, claudin-5), cellular adhesion molecules (ICAM-1, VCAM-1), and endothelial surface markers (EPCR) were determined using immunofluorescence imaging (IF) and western blotting (WB). Expression of angiogenic and cell stress markers were measured using multiplex proteome profiler arrays. RESULTS After 6-h of co-culture with Pf-iRBCs, hiPSC-BMECs showed reduced TEER and increased sodium fluorescein permeability compared to co-culture with uninfected RBCs, indicative of a leaky barrier. We observed disruptions in localization of occludin, zonula occludens-1, and claudin-5 by IF, but no change in protein expression by WB in Pf-iRBC co-cultures. Expression of ICAM-1 and VCAM-1 but not EPCR was elevated in hiPSC-BMECs with Pf-iRBC co-culture compared to uninfected RBC co-culture. In addition, there was an increase in expression of angiogenin, platelet factor-4, and phospho-heat shock protein-27 in the Pf-iRBCs co-culture compared to uninfected RBC co-culture. CONCLUSION These findings demonstrate the validity of our hiPSC-BMECs based model of the BBB, that displays enhanced barrier integrity and appropriate TJ protein localization. In the hiPSC-BMEC co-culture with Pf-iRBCs, reduced TEER, increased paracellular permeability, changes in TJ protein localization, increase in expression of adhesion molecules, and markers of angiogenesis and cellular stress all point towards a novel model with enhanced barrier properties, suitable for investigating pathogenic mechanisms underlying BBB disruption in CM.
Collapse
Affiliation(s)
- Adnan Gopinadhan
- Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, R4-402D 1044 W. Walnut St., Indianapolis, IN, 46202, USA
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jason M Hughes
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, 620 Chestnut Street, Terre Haute, IN, 47809, USA
| | - Andrea L Conroy
- Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, R4-402D 1044 W. Walnut St., Indianapolis, IN, 46202, USA
| | - Chandy C John
- Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, R4-402D 1044 W. Walnut St., Indianapolis, IN, 46202, USA
| | - Scott G Canfield
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, 620 Chestnut Street, Terre Haute, IN, 47809, USA.
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - Dibyadyuti Datta
- Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, R4-402D 1044 W. Walnut St., Indianapolis, IN, 46202, USA.
| |
Collapse
|
7
|
Zhou Y, Huang Y, Yang C, Zang X, Deng H, Liu J, Zhao E, Tian T, Pan L, Xue X. The pathways and the mechanisms by which Cryptococcus enters the brain. Mycology 2024; 15:345-359. [PMID: 39247889 PMCID: PMC11376299 DOI: 10.1080/21501203.2023.2295409] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 12/11/2023] [Indexed: 09/10/2024] Open
Abstract
Generally, Cryptococcus initially infects the respiratory tract, but can spread, eventually crossing the blood-brain barrier (BBB) and causing meningitis or meningoencephalitis. Specifically, Cryptococcus invades the vascular endothelial cells of the BBB, from which it enters the brain. The main mechanisms through which Cryptococcus crosses the BBB are transcellular traversal, the paracellular pathway, and via Trojan horse. In this paper, the mechanisms by which Cryptococcus crosses the BBB were explained in detail. In addition to pathways of entry to the brain, this paper presents a discussion on some rare cryptococcal infections and provides some insights for future research directions.
Collapse
Affiliation(s)
- Yangyu Zhou
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yemei Huang
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Chen Yang
- Department of Respiratory and Critical Care, Weifang Medical College, Weifang, China
| | - Xuelei Zang
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Hengyu Deng
- Department of Laboratory Medicine, Chinese PLA General Hospital, the First Medical Centre, Beijing, China
| | - Jing Liu
- Department of Laboratory Medicine, Chinese PLA General Hospital, the First Medical Centre, Beijing, China
| | - Enqi Zhao
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Tingyue Tian
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Lei Pan
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xinying Xue
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Department of Laboratory Medicine, Chinese PLA General Hospital, the First Medical Centre, Beijing, China
| |
Collapse
|
8
|
Lanser DM, Bennett AB, Vu K, Gelli A. Macropinocytosis as a potential mechanism driving neurotropism of Cryptococcus neoformans. Front Cell Infect Microbiol 2023; 13:1331429. [PMID: 38149006 PMCID: PMC10750359 DOI: 10.3389/fcimb.2023.1331429] [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: 11/01/2023] [Accepted: 11/27/2023] [Indexed: 12/28/2023] Open
Abstract
Cryptococcus neoformans can invade the central nervous system by crossing the blood-brain barrier via a transcellular mechanism that relies on multiple host factors. In this narrative, we review the evidence that a direct interplay between C. neoformans and brain endothelial cells forms the basis for invasion and transmigration across the brain endothelium. Adherence and internalization of C. neoformans is dependent on transmembrane proteins, including a hyaluronic acid receptor and an ephrin receptor tyrosine kinase. We consider the role of EphA2 in facilitating the invasion of the central nervous system by C. neoformans and highlight experimental evidence supporting macropinocytosis as a potential mechanism of internalization and transcytosis. How macropinocytosis might be conclusively demonstrated in the context of C. neoformans is also discussed.
Collapse
Affiliation(s)
| | | | | | - Angie Gelli
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
9
|
Alanazi AH, Chastain DB, Rudraraju M, Parvathagiri V, Shan S, Lin X, Henao-Martínez AF, Franco-Paredes C, Narayanan SP, Somanath PR. A multi-arm, parallel, preclinical study investigating the potential benefits of acetazolamide, candesartan, and triciribine in combination with fluconazole for the treatment of cryptococcal meningoencephalitis. Eur J Pharmacol 2023; 960:176177. [PMID: 37931839 PMCID: PMC10985624 DOI: 10.1016/j.ejphar.2023.176177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/25/2023] [Accepted: 11/01/2023] [Indexed: 11/08/2023]
Abstract
Cryptococcus neoformans, an opportunistic fungal pathogen, primarily infects immunodeficient patients frequently causing cryptococcal meningoencephalitis (CM). Increased intracranial pressure (ICP) is a serious complication responsible for increased morbidity and mortality in CM patients. Non-invasive pharmacological agents that mitigate ICP could be beneficial in treating CM patients. The objective of the study was to investigate the efficacy of acetazolamide (AZA), candesartan (CAN), and triciribine (TCBN), in combination with the antifungal fluconazole, on C. neoformans-induced endothelial, brain, and lung injury in an experimental mouse model of CM. Our study shows that C. neoformans increases the expression of brain endothelial cell (BEC) junction proteins Claudin-5 (Cldn5) and VE-Cadherin to induce pathological cell-barrier remodeling and gap formation associated with increased Akt and p38 MAPK activation. All three agents inhibited C. neoformans-induced endothelial gap formation, only CAN and TCBN significantly reduced C. neoformans-induced Cldn5 expression, and only TCBN was effective in inhibiting Akt and p38MAPK. Interestingly, although C. neoformans did not cause brain or lung edema in mice, it induced lung and brain injuries, which were significantly reversed by AZA, CAN, or TCBN. Our study provides novel insights into the direct effects of C. neoformans on BECs in vitro, and the potential benefits of using AZA, CAN, or TCBN in the management of CM patients.
Collapse
Affiliation(s)
- Abdulaziz H Alanazi
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, 30907, USA; Research Department, Charlie Norwood VA Medical Center, Augusta, GA, 30901, USA
| | - Daniel B Chastain
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, SWGA Clinical Campus, Phoebe Putney Memorial Hospital, Albany, GA, 31701, USA
| | - Madhuri Rudraraju
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, 30907, USA; Research Department, Charlie Norwood VA Medical Center, Augusta, GA, 30901, USA
| | - Varun Parvathagiri
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, 30907, USA; Research Department, Charlie Norwood VA Medical Center, Augusta, GA, 30901, USA
| | - Shengshuai Shan
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, 30907, USA; Research Department, Charlie Norwood VA Medical Center, Augusta, GA, 30901, USA
| | - Xiaorong Lin
- Department of Microbiology, University of Georgia, Athens, GA, 30602, USA
| | - Andrés F Henao-Martínez
- Division of Infectious Diseases, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Carlos Franco-Paredes
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, 80523, USA; Hospital Infantil de México, Federico Gómez, México City, 06720, Mexico
| | - S Priya Narayanan
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, 30907, USA; Research Department, Charlie Norwood VA Medical Center, Augusta, GA, 30901, USA
| | - Payaningal R Somanath
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, 30907, USA; Research Department, Charlie Norwood VA Medical Center, Augusta, GA, 30901, USA.
| |
Collapse
|
10
|
Reguera-Gomez M, Dores MR, Martinez LR. Innovative and potential treatments for fungal central nervous system infections. Curr Opin Microbiol 2023; 76:102397. [PMID: 37898052 DOI: 10.1016/j.mib.2023.102397] [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: 07/31/2023] [Revised: 09/12/2023] [Accepted: 09/25/2023] [Indexed: 10/30/2023]
Abstract
Fungal infections of the central nervous system (FI-CNS) are a problematic and important medical challenge considering that those most affected are immunocompromised. Individuals with systemic cryptococcosis (67-84%), candidiasis (3-64%), blastomycosis (40%), coccidioidomycosis (25%), histoplasmosis (5-20%), mucormycosis (12%), and aspergillosis (4-6%) are highly susceptible to develop CNS involvement, which often results in high mortality (15-100%) depending on the mycosis and the affected immunosuppressed population. Current antifungal drugs are limited, prone to resistance, present host toxicity, and show reduced brain penetration, making FI-CNS very difficult to treat. Given these limitations and the rise in FI-CNS, there is a need for innovative strategies for therapeutic development and treatments to manage FI-CNS in at-risk populations. Here, we discuss standards of care, antifungal drug candidates, and novel molecular targets in the blood-brain barrier, which is a protective structure that regulates movement of particles in and out of the brain, to prevent and combat FI-CNS.
Collapse
Affiliation(s)
- Marta Reguera-Gomez
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL, USA
| | - Michael R Dores
- Department of Biology, Hofstra University, Hempstead, NY, USA
| | - Luis R Martinez
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL, USA; Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA; Center for Immunology and Transplantation, Gainesville, FL, USA; Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
11
|
Lee HH, Carmichael DJ, Ríbeiro V, Parisi DN, Munzen ME, Charles-Niño CL, Hamed MF, Kaur E, Mishra A, Patel J, Rooklin RB, Sher A, Carrillo-Sepulveda MA, Eugenin EA, Dores MR, Martinez LR. Glucuronoxylomannan intranasal challenge prior to Cryptococcus neoformans pulmonary infection enhances cerebral cryptococcosis in rodents. PLoS Pathog 2023; 19:e1010941. [PMID: 37115795 PMCID: PMC10171644 DOI: 10.1371/journal.ppat.1010941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 05/10/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
The encapsulated fungus Cryptococcus neoformans is the most common cause of fungal meningitis, with the highest rate of disease in patients with AIDS or immunosuppression. This microbe enters the human body via inhalation of infectious particles. C. neoformans capsular polysaccharide, in which the major component is glucuronoxylomannan (GXM), extensively accumulates in tissues and compromises host immune responses. C. neoformans travels from the lungs to the bloodstream and crosses to the brain via transcytosis, paracytosis, or inside of phagocytes using a "Trojan horse" mechanism. The fungus causes life-threatening meningoencephalitis with high mortality rates. Hence, we investigated the impact of intranasal exogenous GXM administration on C. neoformans infection in C57BL/6 mice. GXM enhances cryptococcal pulmonary infection and facilitates fungal systemic dissemination and brain invasion. Pre-challenge of GXM results in detection of the polysaccharide in lungs, serum, and surprisingly brain, the latter likely reached through the nasal cavity. GXM significantly alters endothelial cell tight junction protein expression in vivo, suggesting significant implications for the C. neoformans mechanisms of brain invasion. Using a microtiter transwell system, we showed that GXM disrupts the trans-endothelial electrical resistance, weakening human brain endothelial cell monolayers co-cultured with pericytes, supportive cells of blood vessels/capillaries found in the blood-brain barrier (BBB) to promote C. neoformans BBB penetration. Our findings should be considered in the development of therapeutics to combat the devastating complications of cryptococcosis that results in an estimated ~200,000 deaths worldwide each year.
Collapse
Affiliation(s)
- Hiu Ham Lee
- Department of Biomedical Sciences, NYIT College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, New York, United States of America
| | - Dylan J Carmichael
- Department of Biomedical Sciences, NYIT College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, New York, United States of America
| | - Victoria Ríbeiro
- Department of Neuroscience, Cell Biology, and Anatomy, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Dana N Parisi
- Department of Biomedical Sciences, NYIT College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, New York, United States of America
- Department of Biomedical Sciences, Long Island University-Post, Brookville, New York, United States of America
| | - Melissa E Munzen
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, United States of America
| | - Claudia L Charles-Niño
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, United States of America
| | - Mohamed F Hamed
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, United States of America
- Department of Pathology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Ettiman Kaur
- Department of Biology, Hofstra University, Hempstead, New York, United States of America
| | - Ayush Mishra
- Department of Biology, Hofstra University, Hempstead, New York, United States of America
| | - Jiya Patel
- Department of Biology, Hofstra University, Hempstead, New York, United States of America
| | - Rikki B Rooklin
- Department of Biology, Hofstra University, Hempstead, New York, United States of America
| | - Amina Sher
- Department of Biology, Hofstra University, Hempstead, New York, United States of America
| | - Maria A Carrillo-Sepulveda
- Department of Biomedical Sciences, NYIT College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, New York, United States of America
| | - Eliseo A Eugenin
- Department of Neuroscience, Cell Biology, and Anatomy, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Michael R Dores
- Department of Biology, Hofstra University, Hempstead, New York, United States of America
| | - Luis R Martinez
- Department of Biomedical Sciences, NYIT College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, New York, United States of America
- Department of Biomedical Sciences, Long Island University-Post, Brookville, New York, United States of America
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, United States of America
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, United States of America
- Center for Immunology and Transplantation, University of Florida, Gainesville, Florida, United States of America
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, Florida, United States of America
| |
Collapse
|
12
|
Nielson JA, Davis JM. Roles for Microglia in Cryptococcal Brain Dissemination in the Zebrafish Larva. Microbiol Spectr 2023; 11:e0431522. [PMID: 36719205 PMCID: PMC10100726 DOI: 10.1128/spectrum.04315-22] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 12/20/2022] [Indexed: 02/01/2023] Open
Abstract
Cryptococcal infection begins in the lungs, but yeast cells subsequently access the bloodstream, from which they can reach the central nervous system (CNS). The resulting meningoencephalitis is the most common presentation and is very difficult to treat. How this fungus interacts with the blood-brain barrier (BBB) and establishes growth in the brain parenchyma remains a central question in fungal pathogenesis. We and others have developed the zebrafish larva as a model host for cryptococcosis and demonstrated that hematogenous CNS infection is replicated in this model. Here, we have used this model to examine the details of BBB crossing and the events immediately before and after. We have observed multiple mechanisms of BBB crossing and found that microglia, the resident phagocytes of the brain, likely have multiple roles. First, microglia either actively transfer yeast cells across the BBB or take up a significant proportion of them immediately after crossing. Second, microglia are capable of clearing individual cryptococcal cells at a developmental stage before adaptive immune cells have emerged. Third, microglia serve to maintain endothelial integrity, preventing other, phagocyte-independent forms of crossing. These proposed microglial functions during infection in the zebrafish larva generate new hypotheses concerning the establishment and control of cryptococcal meningoencephalitis. IMPORTANCE Cryptococcal meningitis is a fungal infection of the brain and a major cause of death in people with uncontrolled HIV. Infection begins in the lungs but can enter the bloodstream and disseminate to the brain. A structure called the blood-brain barrier must be crossed for the fungus to enter and cause meningitis. Learning how Cryptococcus crosses the blood-brain barrier will be crucial to understanding and possibly preventing brain infection. Using the zebrafish larva as a model host, we show that microglia, the resident phagocytes of the brain, potentially play multiple previously unappreciated roles in cryptococcal infection of the brain. These roles include reinforcing the integrity of the blood-brain barrier, clearing cryptococcal cells after they have crossed, and possibly participating directly in crossing via a previously unknown mechanism.
Collapse
Affiliation(s)
- Jacquelyn A. Nielson
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, USA
| | - J. Muse Davis
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, USA
| |
Collapse
|
13
|
Hamed MF, Enriquez V, Munzen ME, Charles-Niño CL, Mihu MR, Khoshbouei H, Alviña K, Martinez LR. Clinical and pathological characterization of Central Nervous System cryptococcosis in an experimental mouse model of stereotaxic intracerebral infection. PLoS Negl Trop Dis 2023; 17:e0011068. [PMID: 36656900 PMCID: PMC9888703 DOI: 10.1371/journal.pntd.0011068] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/31/2023] [Accepted: 01/02/2023] [Indexed: 01/20/2023] Open
Abstract
Infection of the Central Nervous System (CNS) by the encapsulated fungus Cryptococcus neoformans can lead to high mortality meningitis, most commonly in immunocompromised patients. While the mechanisms by which the fungus crosses the blood-brain barrier to initiate infection in the CNS are well recognized, there are still substantial unanswered questions about the disease progression once the fungus is established in the brain. C. neoformans is characterized by a glucuronoxylomannan (GXM)-rich polysaccharide capsule which has been implicated in immune evasion, but its role during the host CNS infection needs further elucidation. Therefore, the present study aims to examine these key questions about the mechanisms underlying cryptococcal meningitis progression and the impact of fungal GXM release by using an intracerebral rodent infection model via stereotaxic surgery. After developing brain infection, we analyzed distinct brain regions and found that while fungal load and brain weight were comparable one-week post-infection, there were region-specific histopathological (with and without brain parenchyma involvement) and disease manifestations. Moreover, we also observed a region-specific correlation between GXM accumulation and glial cell recruitment. Furthermore, mortality was associated with the presence of subarachnoid hemorrhaging and GXM deposition in the meningeal blood vessels and meninges in all regions infected. Our results show that using the present infection model can facilitate clinical and neuropathological observations during the progression of neurocryptococcosis. Importantly, this mouse model can be used to further investigate disease progression as it develops in humans.
Collapse
Affiliation(s)
- Mohamed F. Hamed
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, United States of America
- Department of Pathology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Vanessa Enriquez
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, United States of America
| | - Melissa E. Munzen
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, United States of America
| | - Claudia L. Charles-Niño
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, United States of America
- Department of Microbiology and Pathology, University Health Sciences Center, University of Guadalajara, Guadalajara, Mexico
| | - Mircea Radu Mihu
- Advanced Critical Care, Nazih Zuhdi Transplant Institute, Advanced Cardiac Care and 24/7 Shock Service, Integris Baptist Medical Center, Oklahoma City, Oklahoma, United States of America
- Department of Medicine/Cardiology, Oklahoma State University Health Science Center, Tulsa, Oklahoma, United States of America
| | - Habibeh Khoshbouei
- Department of Neuroscience, University of Florida, Gainesville, Florida, United States of America
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, United States of America
| | - Karina Alviña
- Department of Neuroscience, University of Florida, Gainesville, Florida, United States of America
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, United States of America
| | - Luis R. Martinez
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, United States of America
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, United States of America
- Center for Immunology and Transplantation, University of Florida, Gainesville, United States of America
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, United States of America
- * E-mail:
| |
Collapse
|
14
|
Li H, Han X, Du W, Meng Y, Li Y, Sun T, Liang Q, Li C, Suo C, Gao X, Qiu Y, Tian W, An M, Zhang H, Fu Y, Li X, Lan T, Yang S, Zhang Z, Geng W, Ding C, Shang H. Comparative miRNA transcriptomics of macaques and mice reveals MYOC is an inhibitor for Cryptococcus neoformans invasion into the brain. Emerg Microbes Infect 2022; 11:1572-1585. [PMID: 35621025 PMCID: PMC9176638 DOI: 10.1080/22221751.2022.2081619] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Cryptococcal meningoencephalitis (CM) is emerging as an infection in HIV/AIDS patients shifted from primarily ARTnaive to ART-experienced individuals, as well as patients with COVID-19 and immunocompetent hosts. This fungal infection is mainly caused by the opportunistic human pathogen Cryptococcus neoformans. Brain or central nervous system (CNS) dissemination is the deadliest process for this disease; however, mechanisms underlying this process have yet to be elucidated. Moreover, illustrations of clinically relevant responses in cryptococcosis are currently limited due to the low availability of clinical samples. In this study, to explore the clinically relevant responses during C. neoformans infection, macaque and mouse infection models were employed and miRNA-mRNA transcriptomes were performed and combined, which revealed cytoskeleton, a major feature of HIV/AIDS patients, was a centric pathway regulated in both infection models. Notably, assays of clinical immune cells confirmed an enhanced macrophage “Trojan Horse” in patients with HIV/AIDS, which could be shut down by cytoskeleton inhibitors. Furthermore, myocilin, encoded by MYOC, was found to be a novel enhancer for the macrophage “Trojan Horse,” and an enhanced fungal burden was achieved in the brains of MYOC-transgenic mice. Taken together, the findings from this study reveal fundamental roles of the cytoskeleton and MYOC in fungal CNS dissemination, which not only helps to understand the high prevalence of CM in HIV/AIDS but also facilitates the development of novel therapeutics for meningoencephalitis caused by C. neoformans and other pathogenic microorganisms.
Collapse
Affiliation(s)
- Hailong Li
- NHC Key Laboratory of AIDS Immunology, National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, People's Republic of China
| | - Xiaoxu Han
- NHC Key Laboratory of AIDS Immunology, National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, People's Republic of China
| | - Wei Du
- College of Life and Health Sciences, Northeastern University, Shenyang, People's Republic of China
| | - Yang Meng
- College of Life and Health Sciences, Northeastern University, Shenyang, People's Republic of China
| | - Yanjian Li
- College of Life and Health Sciences, Northeastern University, Shenyang, People's Republic of China
| | - Tianshu Sun
- Medical Research Centre, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, People's Republic of China.,Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, People's Republic of China
| | - Qiaojing Liang
- College of Life and Health Sciences, Northeastern University, Shenyang, People's Republic of China
| | - Chao Li
- College of Life and Health Sciences, Northeastern University, Shenyang, People's Republic of China
| | - Chenhao Suo
- College of Life and Health Sciences, Northeastern University, Shenyang, People's Republic of China
| | - Xindi Gao
- College of Life and Health Sciences, Northeastern University, Shenyang, People's Republic of China
| | - Yu Qiu
- NHC Key Laboratory of AIDS Immunology, National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, People's Republic of China
| | - Wen Tian
- NHC Key Laboratory of AIDS Immunology, National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, People's Republic of China
| | - Minghui An
- NHC Key Laboratory of AIDS Immunology, National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, People's Republic of China
| | - Hui Zhang
- NHC Key Laboratory of AIDS Immunology, National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, People's Republic of China
| | - Yajing Fu
- NHC Key Laboratory of AIDS Immunology, National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, People's Republic of China
| | - Xiaolin Li
- NHC Key Laboratory of AIDS Immunology, National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, People's Republic of China
| | - Tian Lan
- College of Life and Health Sciences, Northeastern University, Shenyang, People's Republic of China
| | - Sheng Yang
- College of Life and Health Sciences, Northeastern University, Shenyang, People's Republic of China
| | - Zining Zhang
- NHC Key Laboratory of AIDS Immunology, National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, People's Republic of China
| | - Wenqing Geng
- NHC Key Laboratory of AIDS Immunology, National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, People's Republic of China
| | - Chen Ding
- College of Life and Health Sciences, Northeastern University, Shenyang, People's Republic of China
| | - Hong Shang
- NHC Key Laboratory of AIDS Immunology, National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, People's Republic of China
| |
Collapse
|
15
|
Denham ST, Brammer B, Chung KY, Wambaugh MA, Bednarek JM, Guo L, Moreau CT, Brown JCS. A dissemination-prone morphotype enhances extrapulmonary organ entry by Cryptococcus neoformans. Cell Host Microbe 2022; 30:1382-1400.e8. [PMID: 36099922 PMCID: PMC9588642 DOI: 10.1016/j.chom.2022.08.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/01/2022] [Accepted: 08/18/2022] [Indexed: 01/08/2023]
Abstract
Environmental pathogens move from ecological niches to mammalian hosts, requiring adaptation to dramatically different environments. Microbes that disseminate farther, including the fungal meningitis pathogen Cryptococcus neoformans, require additional adaptation to diverse tissues. We demonstrate that the formation of a small C. neoformans morphotype-called "seed" cells due to their colonizing ability-is critical for extrapulmonary organ entry. Seed cells exhibit changes in fungal cell size and surface expression that result in an enhanced macrophage update. Seed cell formation is triggered by environmental factors, including C. neoformans' environmental niche, and pigeon guano with phosphate plays a central role. Seed cells show the enhanced expression of phosphate acquisition genes, and mutants unable to acquire phosphate fail to adopt the seed cell morphotype. Additionally, phosphate can be released by tissue damage, potentially establishing a feed-forward loop of seed cell formation and dissemination. Thus, C. neoformans' size variation represent inducible morphotypes that change host interactions to facilitate microbe spread.
Collapse
Affiliation(s)
- Steven T Denham
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - Brianna Brammer
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - Krystal Y Chung
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - Morgan A Wambaugh
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - Joseph M Bednarek
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - Li Guo
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
| | - Christian T Moreau
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - Jessica C S Brown
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
16
|
Cryptococcus neoformans Infection in the Central Nervous System: The Battle between Host and Pathogen. J Fungi (Basel) 2022; 8:jof8101069. [PMID: 36294634 PMCID: PMC9605252 DOI: 10.3390/jof8101069] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/28/2022] [Accepted: 10/07/2022] [Indexed: 11/17/2022] Open
Abstract
Cryptococcus neoformans (C. neoformans) is a pathogenic fungus with a global distribution. Humans become infected by inhaling the fungus from the environment, and the fungus initially colonizes the lungs. If the immune system fails to contain C. neoformans in the lungs, the fungus can disseminate to the blood and invade the central nervous system, resulting in fatal meningoencephalitis particularly in immunocompromised individuals including HIV/AIDS patients. Following brain invasion, C. neoformans will encounter host defenses involving resident as well as recruited immune cells in the brain. To overcome host defenses, C. neoformans possesses multiple virulence factors capable of modulating immune responses. The outcome of the interactions between the host and C. neoformans will determine the disease progression. In this review, we describe the current understanding of how C. neoformans migrates to the brain across the blood–brain barrier, and how the host immune system responds to the invading organism in the brain. We will also discuss the virulence factors that C. neoformans uses to modulate host immune responses.
Collapse
|
17
|
Reyes EY, Shinohara ML. Host immune responses in the central nervous system during fungal infections. Immunol Rev 2022; 311:50-74. [PMID: 35672656 PMCID: PMC9489659 DOI: 10.1111/imr.13101] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 04/24/2022] [Accepted: 05/18/2022] [Indexed: 12/19/2023]
Abstract
Fungal infections in the central nervous system (CNS) cause high morbidity and mortality. The frequency of CNS mycosis has increased over the last two decades as more individuals go through immunocompromised conditions for various reasons. Nevertheless, options for clinical interventions for CNS mycoses are still limited. Thus, there is an urgent need to understand the host-pathogen interaction mechanisms in CNS mycoses for developing novel treatments. Although the CNS has been regarded as an immune-privileged site, recent studies demonstrate the critical involvement of immune responses elicited by CNS-resident and CNS-infiltrated cells during fungal infections. In this review, we discuss mechanisms of fungal invasion in the CNS, fungal pathogen detection by CNS-resident cells (microglia, astrocytes, oligodendrocytes, neurons), roles of CNS-infiltrated leukocytes, and host immune responses. We consider that understanding host immune responses in the CNS is crucial for endeavors to develop treatments for CNS mycosis.
Collapse
Affiliation(s)
- Estefany Y. Reyes
- Department of Immunology, Duke University School of Medicine, Durham, NC 27705, USA
| | - Mari L. Shinohara
- Department of Immunology, Duke University School of Medicine, Durham, NC 27705, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27705, USA
| |
Collapse
|
18
|
Paredes-Rojas A, Palma-Ramos A, Castrillón-Rivera LE, Mendoza-Pérez F, Navarro-González MDC, Arenas-Guzmán R, Castañeda-Sánchez JI, Luna-Herrera J. Keratinocyte Response to Infection with Sporothrix schenckii. J Fungi (Basel) 2022; 8:jof8050437. [PMID: 35628694 PMCID: PMC9143681 DOI: 10.3390/jof8050437] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 12/27/2022] Open
Abstract
Sporotrichosis is a subacute, or chronic mycosis caused by traumatic inoculation of material contaminated with the fungus Sporothrix schenckii which is part of the Sporothrix spp. complex. The infection is limited to the skin, although its progression to more severe systemic or disseminated forms remains possible. Skin is the tissue that comes into contact with Sporothrix first, and the role of various cell lines has been described with regard to infection control. However, there is little information on the response of keratinocytes. In this study, we used the human keratinocyte cell line (HaCaT) and evaluated different aspects of infection from modifications in the cytoskeleton to the expression of molecules of the innate response during infection with conidia and yeast cells of Sporothrix schenckii. We found that during infection with both phases of the fungus, alterations of the actin cytoskeleton, formation of membrane protuberances, and loss of stress fibers were induced. We also observed an overexpression of the surface receptors MR, TLR6, CR3 and TLR2. Cytokine analysis showed that both phases of the fungus induced the production of elevated levels of the chemokines MCP-1 and IL-8, and proinflammatory cytokines IFN-α, IFN-γ and IL-6. In contrast, TNF-α production was significant only with conidial infection. In late post-infection, cytokine production was observed with immunoregulatory activity, IL-10, and growth factors, G-CSF and GM-CSF. In conclusion, infection of keratinocytes with conidia and yeast cells of Sporothrix schenckii induces an inflammatory response and rearrangements of the cytoskeleton.
Collapse
Affiliation(s)
- Araceli Paredes-Rojas
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico;
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco, Mexico City 04960, Mexico; (A.P.-R.); (L.E.C.-R.); (F.M.-P.)
| | - Alejandro Palma-Ramos
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco, Mexico City 04960, Mexico; (A.P.-R.); (L.E.C.-R.); (F.M.-P.)
| | - Laura Estela Castrillón-Rivera
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco, Mexico City 04960, Mexico; (A.P.-R.); (L.E.C.-R.); (F.M.-P.)
| | - Felipe Mendoza-Pérez
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco, Mexico City 04960, Mexico; (A.P.-R.); (L.E.C.-R.); (F.M.-P.)
| | - María del Carmen Navarro-González
- Laboratorio de Investigación en Enfermedades Reumáticas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City 14080, Mexico;
| | - Roberto Arenas-Guzmán
- Sección de Micología, Hospital General “Dr. Manuel Gea González”, Mexico City 14080, Mexico;
| | - Jorge Ismael Castañeda-Sánchez
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco, Mexico City 04960, Mexico; (A.P.-R.); (L.E.C.-R.); (F.M.-P.)
- Correspondence: (J.I.C.-S.); (J.L.-H.); Tel.: +52-55-54-83-70-00 (ext. 2803) (J.I.C.-S.); +52-55-57-29-63-00 (ext. 62371) (J.L.-H.)
| | - Julieta Luna-Herrera
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico;
- Correspondence: (J.I.C.-S.); (J.L.-H.); Tel.: +52-55-54-83-70-00 (ext. 2803) (J.I.C.-S.); +52-55-57-29-63-00 (ext. 62371) (J.L.-H.)
| |
Collapse
|
19
|
Rathore SS, Sathiyamoorthy J, Lalitha C, Ramakrishnan J. A holistic review on Cryptococcus neoformans. Microb Pathog 2022; 166:105521. [DOI: 10.1016/j.micpath.2022.105521] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 04/06/2022] [Accepted: 04/06/2022] [Indexed: 12/21/2022]
|
20
|
Gutierrez-Gongora D, Geddes-McAlister J. Peptidases: promising antifungal targets of the human fungal pathogen, Cryptococcus neoformans. Facets (Ott) 2022. [DOI: 10.1139/facets-2021-0157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cryptococcus neoformans is a globally important fungal pathogen, primarily inflicting disease on immunocompromised individuals. The widespread use of antifungal agents in medicine and agriculture supports the development of antifungal resistance through evolution, and the emergence of new strains with intrinsic resistance drives the need for new therapeutics. For C. neoformans, the production of virulence factors, including extracellular peptidases (e.g., CnMpr-1 and May1) with mechanistic roles in tissue invasion and fungal survival, constitute approximately 2% of the fungal proteome and cover five classes of enzymes. Given their role in fungal virulence, peptidases represent promising targets for anti-virulence discovery in the development of new approaches against C. neoformans. Additionally, intracellular peptidases, which are involved in resistance mechanisms against current treatment options (e.g., azole drugs), as well as capsule biosynthesis and elaboration of virulence factors, present additional opportunities to combat the pathogen. In this review, we highlight key cryptococcal peptidases with defined or predicted roles in fungal virulence and assess sequence alignments against their human homologs. With this information, we define the feasibility of the select peptidases as “druggable” targets for inhibition, representing prospective therapeutic options against the deadly fungus.
Collapse
Affiliation(s)
- Davier Gutierrez-Gongora
- The Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
- Centro de Estudio de Proteínas, Facultad de Biología, Universidad de La Habana, La Habana, Cuba
| | - Jennifer Geddes-McAlister
- The Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
- Canadian Proteomics and Artificial Intelligence Research and Training Consortium
| |
Collapse
|
21
|
Chen Y, Li C, Sun D, Strickland AB, Liu G, Shi M. Quantitative analysis reveals internalisation of Cryptococcus neoformans by brain endothelial cells in vivo. Cell Microbiol 2021; 23:e13330. [PMID: 33745221 DOI: 10.1111/cmi.13330] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/16/2021] [Accepted: 03/17/2021] [Indexed: 12/12/2022]
Abstract
Migration of Cryptococcus neoformans from the blood to the brain parenchyma is crucial to cause fatal meningoencephalitis. Although mechanisms involved in brain migration of C. neoformans have been widely studied in vitro, less is known about how the fungus crosses the blood-brain barrier (BBB) in vivo. This is in part because of the lack of an approach to quantitatively analyse the dynamics of fungal transmigration into the brain across the BBB in vivo. In this study, we report a novel approach to quantitatively analyse the interactions between C. neoformans and brain endothelial cells in a mouse model using flow cytometry. Using this system, we show that C. neoformans was internalised by brain endothelial cells in vivo and that mice infected with acapsular or heat-killed C. neoformans yeast cells displayed a lower frequency of brain endothelial cells containing the yeast cell compared to mice infected with wild-type or viable yeast cells, respectively. We further demonstrate that brain endothelial cells were invaded by serotype A strain (H99 strain) at a higher rate compared to serotype D strain (52D strain). Our experiments established that internalisation of C. neoformans by brain endothelial cells occurred in vivo and offered a powerful approach to quantitatively analyse fungal migration into the brain.
Collapse
Affiliation(s)
- Yanli Chen
- Division of Immunology, Virginia-Maryland College of Veterinary Medicine and Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA
| | - Chang Li
- Division of Immunology, Virginia-Maryland College of Veterinary Medicine and Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA
| | - Donglei Sun
- Division of Immunology, Virginia-Maryland College of Veterinary Medicine and Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA
| | - Ashley B Strickland
- Division of Immunology, Virginia-Maryland College of Veterinary Medicine and Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA
| | - Gongguan Liu
- Division of Immunology, Virginia-Maryland College of Veterinary Medicine and Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA
| | - Meiqing Shi
- Division of Immunology, Virginia-Maryland College of Veterinary Medicine and Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA
| |
Collapse
|
22
|
Strickland AB, Shi M. Mechanisms of fungal dissemination. Cell Mol Life Sci 2021; 78:3219-3238. [PMID: 33449153 PMCID: PMC8044058 DOI: 10.1007/s00018-020-03736-z] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 11/23/2020] [Accepted: 12/08/2020] [Indexed: 12/13/2022]
Abstract
Fungal infections are an increasing threat to global public health. There are more than six million fungal species worldwide, but less than 1% are known to infect humans. Most of these fungal infections are superficial, affecting the hair, skin and nails, but some species are capable of causing life-threatening diseases. The most common of these include Cryptococcus neoformans, Aspergillus fumigatus and Candida albicans. These fungi are typically innocuous and even constitute a part of the human microbiome, but if these pathogens disseminate throughout the body, they can cause fatal infections which account for more than one million deaths worldwide each year. Thus, systemic dissemination of fungi is a critical step in the development of these deadly infections. In this review, we discuss our current understanding of how fungi disseminate from the initial infection sites to the bloodstream, how immune cells eliminate fungi from circulation and how fungi leave the blood and enter distant organs, highlighting some recent advances and offering some perspectives on future directions.
Collapse
Affiliation(s)
- Ashley B Strickland
- Division of Immunology, Virginia-Maryland College of Veterinary Medicine and Maryland Pathogen Research Institute, University of Maryland, College Park, MD, USA.
| | - Meiqing Shi
- Division of Immunology, Virginia-Maryland College of Veterinary Medicine and Maryland Pathogen Research Institute, University of Maryland, College Park, MD, USA.
| |
Collapse
|
23
|
The Transcription Factor Pdr802 Regulates Titan Cell Formation and Pathogenicity of Cryptococcus neoformans. mBio 2021; 12:mBio.03457-20. [PMID: 33688010 PMCID: PMC8092302 DOI: 10.1128/mbio.03457-20] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The pathogenic yeast Cryptococcus neoformans presents a worldwide threat to human health, especially in the context of immunocompromise, and current antifungal therapy is hindered by cost, limited availability, and inadequate efficacy. After the infectious particle is inhaled, C. neoformans initiates a complex transcriptional program that integrates cellular responses and enables adaptation to the host lung environment. Cryptococcus neoformans is a ubiquitous, opportunistic fungal pathogen that kills almost 200,000 people worldwide each year. It is acquired when mammalian hosts inhale the infectious propagules; these are deposited in the lung and, in the context of immunocompromise, may disseminate to the brain and cause lethal meningoencephalitis. Once inside the host, C. neoformans undergoes a variety of adaptive processes, including secretion of virulence factors, expansion of a polysaccharide capsule that impedes phagocytosis, and the production of giant (Titan) cells. The transcription factor Pdr802 is one regulator of these responses to the host environment. Expression of the corresponding gene is highly induced under host-like conditions in vitro and is critical for C. neoformans dissemination and virulence in a mouse model of infection. Direct targets of Pdr802 include the quorum sensing proteins Pqp1, Opt1, and Liv3; the transcription factors Stb4, Zfc3, and Bzp4, which regulate cryptococcal brain infectivity and capsule thickness; the calcineurin targets Had1 and Crz1, important for cell wall remodeling and C. neoformans virulence; and additional genes related to resistance to host temperature and oxidative stress, and to urease activity. Notably, cryptococci engineered to lack Pdr802 showed a dramatic increase in Titan cells, which are not phagocytosed and have diminished ability to directly cross biological barriers. This explains the limited dissemination of pdr802 mutant cells to the central nervous system and the consequently reduced virulence of this strain. The role of Pdr802 as a negative regulator of Titan cell formation is thus critical for cryptococcal pathogenicity.
Collapse
|
24
|
Araújo GRDS, Alves V, Martins-de-Souza PH, Guimarães AJ, Honorato L, Nimrichter L, Takiya CM, Pontes B, Frases S. Dexamethasone and Methylprednisolone Promote Cell Proliferation, Capsule Enlargement, and in vivo Dissemination of C. neoformans. FRONTIERS IN FUNGAL BIOLOGY 2021; 2:643537. [PMID: 37744119 PMCID: PMC10512211 DOI: 10.3389/ffunb.2021.643537] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 01/19/2021] [Indexed: 09/26/2023]
Abstract
Cryptococcus neoformans is a fungal pathogen that causes life-threatening infections in immunocompromised individuals, who often have some inflammatory condition and, therefore, end up using glucocorticoids, such as dexamethasone and methylprednisolone. Although the effects of this class of molecules during cryptococcosis have been investigated, their consequences for the biology of C. neoformans is less explored. Here, we studied the effects of dexamethasone and methylprednisolone on the metabolism and on the induction of virulence factors in C. neoformans. Our results showed that both glucocorticoids increased fungal cell proliferation and surface electronegativity but reduced capsule and secreted polysaccharide sizes, as well as capsule compaction, by decreasing the density of polysaccharide fibers. We also tested whether glucocorticoids could affect the fungal virulence in Galleria mellonella and mice. Although the survival rate of Galleria larvae increased, those from mice showed a tendency to decrease, with infected animals dying earlier after glucocorticoid treatments. The pathogenesis of spread of cryptococcosis and the interleukin secretion pattern were also assessed for lungs and brains of infected mice. While increases in the spread of the fungus to lungs were observed after treatment with glucocorticoids, a significant difference in brain was observed only for methylprednisolone, although a trend toward increasing was also observed for dexamethasone. Moreover, increases in both pulmonary and cerebral IL-10 production, reduction of IL-6 production but no changes in IL-4, IL-17, and INF-γ were also observed after glucocorticoid treatments. Finally, histopathological analysis confirmed the increase in number of fungal cells in lung and brain tissues of mice previously subjected to dexamethasone or methylprednisolone treatments. Together, our results provide compelling evidence for the effects of dexamethasone and methylprednisolone on the biology of C. neoformans and may have important implications for future clinical treatments, calling attention to the risks of using these glucocorticoids against cryptococcosis or in immunocompromised individuals.
Collapse
Affiliation(s)
- Glauber R. de S. Araújo
- Laboratório de Ultraestrutura Cellular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratório de Biofísica de Fungos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vinicius Alves
- Laboratório de Ultraestrutura Cellular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratório de Biofísica de Fungos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro H. Martins-de-Souza
- Laboratório de Ultraestrutura Cellular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratório de Biofísica de Fungos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Allan J. Guimarães
- Laboratório de Bioquímica e Imunologia das Micoses, Depto. de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Brazil
| | - Leandro Honorato
- Instituto de Microbiologia Paulo de Góes, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leonardo Nimrichter
- Instituto de Microbiologia Paulo de Góes, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Christina Maeda Takiya
- Laboratório de Imunopatologia. Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bruno Pontes
- Instituto de Ciências Biomédicas, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
- Centro Nacional de Biologia Estrutural e Bioimagem (CENABIO), Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Susana Frases
- Laboratório de Ultraestrutura Cellular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratório de Biofísica de Fungos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
25
|
Snarr BD, Drummond RA, Lionakis MS. It's all in your head: antifungal immunity in the brain. Curr Opin Microbiol 2020; 58:41-46. [PMID: 32828989 PMCID: PMC7438209 DOI: 10.1016/j.mib.2020.07.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 12/16/2022]
Abstract
As the incidence rate of invasive fungal infections has increased with the use of modern medical interventions, so too has the occurrence of fungi invading the brain. Fungi such as Candida albicans, Cryptococcus neoformans, and Aspergillus fumigatus often infect immunocompromised individuals, and can use several strategies to invade the central nervous system (CNS) by penetrating the blood-brain barrier. Once in the brain parenchyma the specialized resident immune cells need to effectively recognize the fungus and mount an appropriate immune response to clear the infection, without causing debilitating immune-mediated toxicity and neuronal damage. Here we review the current knowledge pertaining to the antifungal response of the CNS and highlight areas where future research is required.
Collapse
Affiliation(s)
- Brendan D Snarr
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy & Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Rebecca A Drummond
- Institute of Immunology & Immunotherapy, Institute of Microbiology & Infection, University of Birmingham, Birmingham, UK
| | - Michail S Lionakis
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy & Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
26
|
Li W, Yin Y, Meng Y, Zhou H, Ma Z, Lin H, Fan H. Proteomic analysis of bEnd.3 cells infected with wild-type and stk-deficient strains of Streptococcus suis serotype 2 reveals protein and pathway regulation. J Proteomics 2020; 230:103983. [PMID: 32961345 DOI: 10.1016/j.jprot.2020.103983] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/20/2020] [Accepted: 09/08/2020] [Indexed: 12/19/2022]
Abstract
Streptococcus suis serotype 2 (SS2) is a zoonotic pathogen causing meningitis in humans and pigs. However, information on the comparative protein expression of the blood-brain barrier (BBB) following SS2 infection is limited. Deletion of the serine/threonine kinase (stk) gene can decrease the ability of SS2 to invade the BBB. In the present study, bEnd.3 cells were used as the BBB model, and a SILAC comparative quantitative proteomic study of bEnd.3 cells infected with the SS2 ZY05719 or Δstk strain was performed to determine the differences between these strains infections. Compared with ZY05719-infected cells, 241 proteins were highly upregulated, and 81 were significantly downregulated in Δstk-infected cells. The obtained data revealed major changes in the proteins involved in RNA process, host cytoskeleton, tight junction disruption and immune response. Some differentially expressed proteins were screened by quantitative real-time PCR to examine their regulation at the transcriptional level, and western blot analysis was used to validate the changes of some selected proteins at the translational level. The results obtained in this study may be useful to understand the host response to SS2 infection and provide crucial clues to decipher how STK expression in SS2 helps the bacteria penetrate the BBB. SIGNIFICANCE: A SILAC comparative quantitative proteomic assay was performed in bEnd.3 cells infected with the SS2 ZY05719 or Δstk strain. 241 upregulated and 81 downregulated differentially expressed proteins (DEPs) were identified. DEPs are involved in RNA process, host cytoskeleton, tight junction disruption and immune response. Some DEPs were examined by qPCR and western blot assays, which were similar to those of their corresponding proteins in the quantitative proteomics analysis.
Collapse
Affiliation(s)
- Weiyi Li
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yifan Yin
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yu Meng
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Hong Zhou
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Zhe Ma
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Huixing Lin
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Hongjie Fan
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.
| |
Collapse
|
27
|
Gupta S, Paul K, Kaur S. Diverse species in the genus Cryptococcus: Pathogens and their non-pathogenic ancestors. IUBMB Life 2020; 72:2303-2312. [PMID: 32897638 DOI: 10.1002/iub.2377] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/15/2020] [Accepted: 08/16/2020] [Indexed: 12/14/2022]
Abstract
The genus Cryptococcus comprises of more than 30 species. It consists of clinically significant pathogenic Cryptococcus neoformans/Cryptococcus gattii species complex comprising of a minimum of seven species. These pathogens cost more than 200,000 lives annually by causing cryptococcal meningoencephalitis. The evolution of the pathogenic species from closely related non-pathogenic species of the Cryptococcus amylolentus complex is of particular importance and several advances have been made to understand their phylogenetic and genomic relationships. The current review briefly describes the sexual reproduction process followed by an individual description of the members focusing on their key attributes and virulence mechanisms of the pathogenic species. A special section on phylogenetic studies is aimed at understanding the evolutionary divergence of pathogens from non-pathogens. Recent findings from our group pertaining to parameters affecting codon usage bias in six pathogenic and three non-pathogenic ancestral species and their corroboration with existing phylogenetic reports are also included in the current review.
Collapse
Affiliation(s)
- Shelly Gupta
- Department of Biochemistry, Lovely Professional University, Kapurthala, India
| | - Karan Paul
- Department of Biochemistry, DAV University, Jalandhar, India
| | - Sukhmanjot Kaur
- Department of Biochemistry, Lovely Professional University, Kapurthala, India
| |
Collapse
|
28
|
An Antivirulence Approach for Preventing Cryptococcus neoformans from Crossing the Blood-Brain Barrier via Novel Natural Product Inhibitors of a Fungal Metalloprotease. mBio 2020; 11:mBio.01249-20. [PMID: 32694141 PMCID: PMC7374060 DOI: 10.1128/mbio.01249-20] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Fungal infections like cryptococcal meningitis are difficult to resolve because of the limited therapies available. The small arsenal of antifungal drugs reflect the difficulty in finding available targets in fungi because like mammalian cells, fungi are eukaryotes. The limited efficacy, toxicity, and rising resistance of antifungals contribute to the high morbidity and mortality of fungal infections and further underscore the dire but unmet need for new antifungal drugs. The traditional approach in antifungal drug development has been to target fungal growth, but an attractive alternative is to target mechanisms of pathogenesis. An important attribute of Cryptococcus neoformans (Cn) pathogenesis is its ability to enter the central nervous system. Here, we describe a large-scale screen that identified three natural products that prevented Cn from crossing the blood-brain barrier by inhibiting the virulence factor Mpr1 without affecting the growth of Cn. We propose that compounds identified here could be further developed as antivirulence therapy that would be administered preemptively or serve as a prophylactic in patients at high risk for developing cryptococcal meningitis. Cryptococcus neoformans (Cn) is the leading cause of fungal meningitis, a deadly disease with limited therapeutic options. Dissemination to the central nervous system hinges on the ability of Cn to breach the blood-brain barrier (BBB) and is considered an attribute of Cn virulence. Targeting virulence instead of growth for antifungal drug development has not been fully exploited despite the benefits of this approach. Mpr1 is a secreted fungal metalloprotease not required for fungal growth, but rather, it functions as a virulence factor by facilitating Cn migration across the BBB. This central role for Mpr1, its extracellular location, and lack of expression in mammalian cells make Mpr1 a high-value target for an antivirulence approach aimed at developing therapeutics for cryptococcal meningitis. To test this notion, we devised a large-scale screen to identify compounds that prohibited Cn from crossing the BBB by selectively blocking Mpr1 proteolytic activity, without inhibiting the growth of Cn. A phytochemical natural product-derived library was screened to identify new molecular scaffolds of prototypes unique to a Cn microecosystem. Of the 240 pure natural products examined, 3 lead compounds, abietic acid, diosgenin, and lupinine inhibited Mpr1 proteolytic activity with 50% inhibitory concentration (IC50) values of <10 μM, displayed little to no mammalian cell toxicity, and did not affect Cn growth. Notably, the lead compounds blocked Cn from crossing the BBB, without damaging the barrier integrity, suggesting the bioactive molecules had no off-target effects. We propose that these new drug scaffolds are promising candidates for the development of antivirulence therapy against cryptococcal meningitis.
Collapse
|
29
|
Abstract
Among fungal pathogens, Cryptococcus neoformans has gained great importance among the scientific community of several reasons. This fungus is the causative agent of cryptococcosis, a disease mainly associated to HIV immunosuppression and characterized by the appearance of meningoencephalitis. Cryptococcal meningitis is responsible for hundreds of thousands of deaths every year. Research of the pathogenesis and virulence mechanisms of this pathogen has focused on three main different areas: Adaptation to the host environment (nutrients, pH, and free radicals), mechanism of immune evasion (which include phenotypic variations and the ability to behave as a facultative intracellular pathogen), and production of virulence factors. Cryptococcus neoformans has two phenotypic characteristics, the capsule and synthesis of melanin that have a profound effect in the virulence of the yeast because they both have protective effects and induce host damage as virulence factors. Finally, the mechanisms that result in dissemination and brain invasion are also of key importance to understand cryptococcal disease. In this review, I will provide a brief overview of the main mechanisms that makes C. neoformans a pathogen in susceptible patients. Abbreviations: RNS: reactive nitrogen species; BBB: brain blood barrier; GXM: glucuronoxylomannan; GXMGal: glucuronoxylomannogalactan
Collapse
Affiliation(s)
- Oscar Zaragoza
- a Mycology Reference Laboratory National Centre for Microbiology , Instituto de Salud Carlos III Carretera Majadahonda-Pozuelo , Madrid , Spain
| |
Collapse
|
30
|
Abstract
Macrophages are well known for their phagocytic activity and their role in innate immune responses. Macrophages eat non-self particles, via a variety of mechanisms, and typically break down internalized cargo into small macromolecules. However, some pathogenic agents have the ability to evade this endosomal degradation through a nonlytic exocytosis process termed vomocytosis. Macrophages are well known for their phagocytic activity and their role in innate immune responses. Macrophages eat non-self particles, via a variety of mechanisms, and typically break down internalized cargo into small macromolecules. However, some pathogenic agents have the ability to evade this endosomal degradation through a nonlytic exocytosis process termed vomocytosis. This phenomenon has been most often studied for Cryptococcus neoformans, a yeast that causes roughly 180,000 deaths per year, primarily in immunocompromised (e.g., human immunodeficiency virus [HIV]) patients. Existing dogma purports that vomocytosis involves distinctive cellular pathways and intracellular physicochemical cues in the host cell during phagosomal maturation. Moreover, it has been observed that the immunological state of the individual and macrophage phenotype affect vomocytosis outcomes. Here we compile the current knowledge on the factors (with respect to the phagocytic cell) that promote vomocytosis of C. neoformans from macrophages.
Collapse
|
31
|
Liu T, Woo JAA, Yan Y, LePochat P, Bukhari MZ, Kang DE. Dual role of cofilin in APP trafficking and amyloid-β clearance. FASEB J 2019; 33:14234-14247. [PMID: 31646885 DOI: 10.1096/fj.201901268r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The accumulation of amyloid-β (Aβ) plays a pivotal early event in the pathogenesis of Alzheimer's disease (AD). In the brain, neurons produce Aβ by the proteolytic processing of amyloid precursor protein (APP) through the endocytic pathway, whereas microglia mediate Aβ clearance also via endocytic mechanisms. Previous studies have shown the critical importance of cofilin, a filamentous actin-severing protein, in actin dynamics and pathogen-triggered endocytic processes. Moreover, the binding of Aβ42 oligomers to β1-integrin triggers the cofilin activation, and in turn, cofilin promotes the internalization of surface β1-integrin. However, a role for cofilin in APP processing and Aβ metabolism has not been investigated. In this study, we found that knockdown of cofilin in Chinese hamster ovary 7WD10 cells and primary neurons significantly reduces Aβ production by increasing surface APP (sAPP) levels. Expression of active (S3A) but not inactive (S3E) cofilin reduces sAPP levels by enhancing APP endocytosis. Accordingly, Aβ deposition in APP and presenilin 1 (PS1) transgenic mice is significantly reduced by genetic reduction of cofilin (APP/PS1;cofilin+/-). However, the reduction of Aβ load in APP/PS1;cofilin+/- mice is paradoxically associated with significantly increased ionized calcium-binding adaptor molecule 1-positive microglial activation surrounding Aβ deposits. Primary microglia isolated from cofilin+/- mice demonstrate significantly enhanced state of activation and greater ability to uptake and clear Aβ42, which is reversed with the active (S3A) but not inactive (S3E) form of cofilin. These results taken together indicate a significant role for cofilin in Aβ accumulation via dual and opposing endocytic mechanisms of promoting Aβ production in neurons and inhibiting Aβ clearance in microglia.-Liu, T., Woo, J.-A. A., Yan, Y., LePochat, P., Bukhari, M. Z., Kang, D. E. Dual role of cofilin in APP trafficking and amyloid-β clearance.
Collapse
Affiliation(s)
- Tian Liu
- Byrd Alzheimer's Institute, Morsani College of Medicine, University of South Florida (USF) Health, Tampa, Florida, USA.,Department of Molecular of Medicine, Morsani College of Medicine, University of South Florida (USF) Health, Tampa, Florida, USA
| | - Jung-A A Woo
- Byrd Alzheimer's Institute, Morsani College of Medicine, University of South Florida (USF) Health, Tampa, Florida, USA.,Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida (USF) Health, Tampa, Florida, USA
| | - Yan Yan
- Byrd Alzheimer's Institute, Morsani College of Medicine, University of South Florida (USF) Health, Tampa, Florida, USA.,Department of Molecular of Medicine, Morsani College of Medicine, University of South Florida (USF) Health, Tampa, Florida, USA
| | - Patrick LePochat
- Byrd Alzheimer's Institute, Morsani College of Medicine, University of South Florida (USF) Health, Tampa, Florida, USA.,Department of Molecular of Medicine, Morsani College of Medicine, University of South Florida (USF) Health, Tampa, Florida, USA
| | - Mohammed Zaheen Bukhari
- Byrd Alzheimer's Institute, Morsani College of Medicine, University of South Florida (USF) Health, Tampa, Florida, USA.,Department of Molecular of Medicine, Morsani College of Medicine, University of South Florida (USF) Health, Tampa, Florida, USA
| | - David E Kang
- Byrd Alzheimer's Institute, Morsani College of Medicine, University of South Florida (USF) Health, Tampa, Florida, USA.,Department of Molecular of Medicine, Morsani College of Medicine, University of South Florida (USF) Health, Tampa, Florida, USA.,James A. Haley Veterans Administration Hospital, Tampa, Florida, USA
| |
Collapse
|
32
|
Luo W, Liang P, Puthiyakunnon S, Yang L, Hong C. VP1-binding protein glucose-regulated protein 78 as an important mediator for enterovirus 71 infecting human brain microvascular endothelial cells. Indian J Med Microbiol 2019; 37:406-414. [PMID: 32003341 DOI: 10.4103/ijmm.ijmm_19_194] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Purpose Enterovirus 71 (EV71) is one of the main pathogens causing hand, foot and mouth disease, which could even induce severe brain damage in some patients. As the underlying mechanism of the invasion and replication process still remains largely unknown, we investigated the role of candidate proteins expressed during EV71 invasion in human brain microvascular endothelial cells (HBMECs) to delineate the pathophysiological mechanism of EV-71 infection. Materials and Methods Ninety-one candidate EV71-associated proteins which could bind the major capsid protein (viral protein 1 [VP1]) of EV71 on the HBMEC were identified by applying an analysis of glutathione-S-transferase pull-down coupling with liquid chromatography-electrospray ionisation-tandem mass spectrometry (LC-ESI-MS/MS). Seventy-eight kDa glucose-regulated protein 78 (GRP78) binding to the VP1 protein was further validated by co-immunoprecipitation, immunofluorescence and western blot analysis. To explore the role of GRP78 in EV71 infection, GRP78 was knocked down and overexpressed in HBMEC and was verified by TCID50 assay. Results LC-ESI-MS/MS-identified 91 proteins were subjected to gene ontology analysis, and on molecular and biological function analysis revealed GRP78 act as an important binding protein in mediating EV71 infection. In addition, immunofluorescence demonstrated the co-localisation of GRP78 and VP1 in cytoplasm of the infected HBMEC. The TCID50 assay showed that knockdown of GRP78 could attenuate the replication capacity of EV71 in HBMEC, and the overexpression could increase the virus titre in HBEMC at 24 h post-infection suggesting that GRP78 was associated with the replication capacity of EV71 in HBMEC. Conclusion These findings provided evidence that GRP78 plays an important role during the progression of EV71 infection as a mediator in HBMEC.
Collapse
Affiliation(s)
- Wenying Luo
- Department of Clinical Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang; Department of Laboratory Medicine, School of Laboratory Medicine, Guangdong Medical University, Guangdong, China
| | - Peng Liang
- Department of Clinic Laboratory, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Santhosh Puthiyakunnon
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Lawei Yang
- Department of Clinical Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Cao Hong
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| |
Collapse
|
33
|
Zhang C, Chen F, Liu X, Han X, Hu Y, Su X, Chen Y, Sun Y, Han L. Gliotoxin Induces Cofilin Phosphorylation to Promote Actin Cytoskeleton Dynamics and Internalization of Aspergillus fumigatus Into Type II Human Pneumocyte Cells. Front Microbiol 2019; 10:1345. [PMID: 31275272 PMCID: PMC6591310 DOI: 10.3389/fmicb.2019.01345] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 05/29/2019] [Indexed: 01/27/2023] Open
Abstract
Aspergillus fumigatus is able to internalize into lung epithelial cells to escape from immune attack for further dissemination. We previously reported that gliotoxin, a major mycotoxin of A. fumigatus, promotes this internalization; however, the mechanism remained unclear. Here, we report that gliotoxin is able to induce cofilin phosphorylation in A549 type II human pneumocytes. Either too high or too low a level of cofilin phosphorylation blocked the gliotoxin-induced actin cytoskeleton rearrangement and A. fumigatus internalization. LIM domain kinase 1 (LIMK1) and its upstream small GTPases (Cdc42 and RhoA, but not Rac1) predominantly mediated the gliotoxin-induced cofilin phosphorylation and A. fumigatus internalization. Simultaneously, gliotoxin significantly stimulated an increase in cAMP; however, adding an antagonist of PKA did not block gliotoxin-induced A. fumigatus internalization. In vivo, exogenous gliotoxin helped gliotoxin synthesis deficient strain gliPΔ invade into the lung tissue and the lung fungal burden increased markedly in immunosuppressed mice. In conclusion, these data revealed a novel role of gliotoxin in inducing cofilin phosphorylation mostly through the Cdc42/RhoA-LIMK1 signaling pathway to promote actin cytoskeleton rearrangement and internalization of A. fumigatus into type II human pneumocytes.
Collapse
Affiliation(s)
- Changjian Zhang
- Chinese PLA Center for Disease Control and Prevention, Beijing, China.,Academy of Military Medical Sciences, Beijing, China
| | - Fangyan Chen
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Xiaoyu Liu
- Chinese PLA Center for Disease Control and Prevention, Beijing, China.,Academy of Military Medical Sciences, Beijing, China
| | - Xuelin Han
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Yingsong Hu
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Xueting Su
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Yong Chen
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Yansong Sun
- Academy of Military Medical Sciences, Beijing, China
| | - Li Han
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
34
|
Lahiri S, Banerjee A, Bhutda S, Palaniappan M, Bahubali VH, Manjunath N, Maji S, Siddaiah N. In vitro expression of vital virulent genes of clinical and environmental isolates of Cryptococcus neoformans/gattii in endothelial cells of human blood-brain barrier. J Mycol Med 2019; 29:239-244. [PMID: 31221506 DOI: 10.1016/j.mycmed.2019.06.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 06/02/2019] [Accepted: 06/04/2019] [Indexed: 11/28/2022]
Abstract
BACKGROUND Evaluation of the pathogenesis of clinical and environmental cryptococcal isolates to the central nervous system is necessary for understanding the risk. This study was designed to determine the in vitro expression of six important virulent genes of Cryptococcus neoformans/gattii in Human Brain Microvascular Endothelial cells (hBMEC). METHODS The hBMEC were infected with Cryptococcus to determine invasion and survival rate at 3, 12 and 24hours by subsequent colony count of internalized yeasts. The whole RNA of the intracellular Cryptococcus was extracted to quantify the expression of CAP10, PLB1, ENA1, URE1, LAC1, and MATα genes by real-time quantitative PCR for 3 and 12hours of infection. RESULTS Invasion and survival rates were higher in clinical and standard strains of C. neoformans. A significant difference was observed among the clinical and environmental isolates for the expression of CAP10, ENA1, LAC1, MATα and URE1 at 3hours, and ENA1, LAC1, MATα, PLB1 and URE1 at 12hours. Clinical isolates showed significant upregulation of all the genes except PLB1, which was higher in environmental isolates. Relative expressions at the two time-points showed statistically significant (P=0.043) changes for the clinical isolates and no significance (P=0.063) for environmental isolates. CONCLUSION The C. gattii (VGI) isolates showed significantly lower invasion and survival than C. neoformans (VNI, and VNII) irrespective of their sources. Clinical isolates exhibited higher expression for the majority of the virulent genes until 12hours of infection, probably due to their better adaptation in the host system and enhanced pathogenicity than the environmental counterparts.
Collapse
Affiliation(s)
- S Lahiri
- Department of Neuromicrobiology, National Institute of Mental Health & Neuro Sciences (NIMHANS), Bangalore, India
| | - A Banerjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Mumbai, India
| | - S Bhutda
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Mumbai, India
| | - M Palaniappan
- Department of Biostatistics, NIMHANS, Bangalore, India
| | - V H Bahubali
- Department of Neuromicrobiology, National Institute of Mental Health & Neuro Sciences (NIMHANS), Bangalore, India
| | - N Manjunath
- Department of Neurology, NIMHANS, Bangalore, India
| | - S Maji
- Department of Neuromicrobiology, National Institute of Mental Health & Neuro Sciences (NIMHANS), Bangalore, India
| | - N Siddaiah
- Department of Neuromicrobiology, National Institute of Mental Health & Neuro Sciences (NIMHANS), Bangalore, India.
| |
Collapse
|
35
|
de Oliveira HC, Trevijano-Contador N, Garcia-Rodas R. Cryptococcal Pathogenicity and Morphogenesis. CURRENT FUNGAL INFECTION REPORTS 2019. [DOI: 10.1007/s12281-019-00340-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
36
|
Vu K, Garcia JA, Gelli A. Cryptococcal Meningitis and Anti-virulence Therapeutic Strategies. Front Microbiol 2019; 10:353. [PMID: 30863389 PMCID: PMC6399105 DOI: 10.3389/fmicb.2019.00353] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 02/11/2019] [Indexed: 01/31/2023] Open
Abstract
Fungal infections of the central nervous system are responsible for significant morbidity and mortality. Cryptococcus neoformans (Cn) is the primary cause of fungal meningitis. Infection begins in the lung after inhalation of fungal spores but often spreads to other organs, particularly the brain in immunosuppressed individuals. Cn’s ability to survive phagocytosis and endure the onslaught of oxidative attack imposed by the innate immune response facilitates dissemination to the central nervous system (CNS). Despite the success of Cn at bypassing innate immunity, entry into the heavily protected brain requires that Cn overwhelm the highly restricted blood-brain barrier (BBB). This is a formidable task but mounting evidence suggests that Cn expresses surface-bound and secreted virulence factors including urease, metalloprotease, and hyaluronic acid that can undermine the BBB. In addition, Cn can exploit multiple routes of entry to gain access to the CNS. In this review, we discuss the cellular and molecular interface of Cn and the BBB, and we propose that the virulence factors mediating BBB crossing could be targeted for the development of anti-virulence drugs aimed at preventing fungal colonization of the CNS.
Collapse
Affiliation(s)
- Kiem Vu
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Javier A Garcia
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Angie Gelli
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
37
|
Santiago-Tirado FH, Klein RS, Doering TL. An In Vitro Brain Endothelial Model for Studies of Cryptococcal Transmigration into the Central Nervous System. ACTA ACUST UNITED AC 2019; 53:e78. [PMID: 30776307 DOI: 10.1002/cpmc.78] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cryptococcus neoformans is an environmental yeast found worldwide that causes lethal brain infections, particularly in immunocompromised hosts. In 2016, there were 280,000 cases of cryptococcal meningitis in the HIV+ population, two-thirds of them fatal; other immunocompromised patients are also affected. The burden of cryptococcal disease and the limits of current chemotherapy create a pressing need for improved treatment. One hindrance to the development of new therapies is lack of understanding of how this pathogen breaches the barriers protecting the brain. Here we describe a tool for investigating this process. This simple in vitro blood-brain-barrier (BBB) model, based on a human brain endothelial cell line grown on a permeable membrane, may be used to assay the BBB transmigration of C. neoformans or other neurotropic pathogens. © 2019 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Felipe H Santiago-Tirado
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri.,Current address: Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana
| | - Robyn S Klein
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Tamara L Doering
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
38
|
Shourian M, Qureshi ST. Resistance and Tolerance to Cryptococcal Infection: An Intricate Balance That Controls the Development of Disease. Front Immunol 2019; 10:66. [PMID: 30761136 PMCID: PMC6361814 DOI: 10.3389/fimmu.2019.00066] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 01/11/2019] [Indexed: 12/25/2022] Open
Abstract
Cryptococcus neoformans is a ubiquitous environmental yeast and a leading cause of invasive fungal infection in humans. The most recent estimate of global disease burden includes over 200,000 cases of cryptococcal meningitis each year. Cryptococcus neoformans expresses several virulence factors that may have originally evolved to protect against environmental threats, and human infection may be an unintended consequence of these acquired defenses. Traditionally, C. neoformans has been viewed as a purely opportunistic pathogen that targets severely immune compromised hosts; however, during the past decade the spectrum of susceptible individuals has grown considerably. In addition, the closely related strain Cryptococcus gattii has recently emerged in North America and preferentially targets individuals with intact immunity. In parallel to the changing epidemiology of cryptococcosis, an increasing role for host immunity in the pathogenesis of severe disease has been elucidated. Initially, the HIV/AIDS epidemic revealed the capacity of C. neoformans to cause host damage in the absence of adaptive immunity. Subsequently, the development and clinical implementation of highly active antiretroviral treatment (HAART) led to recognition of an immune reconstitution inflammatory syndrome (IRIS) in a subset of HIV+ individuals, demonstrating the pathological role of host immunity in disease. A post-infectious inflammatory syndrome (PIIRS) characterized by abnormal T cell-macrophage activation has also been documented in HIV-negative individuals following antifungal therapy. These novel clinical conditions illustrate the highly complex host-pathogen relationship that underlies severe cryptococcal disease and the intricate balance between tolerance and resistance that is necessary for effective resolution. In this article, we will review current knowledge of the interactions between cryptococci and mammalian hosts that result in a tolerant phenotype. Future investigations in this area have potential for translation into improved therapies for affected individuals.
Collapse
Affiliation(s)
- Mitra Shourian
- Translational Research in Respiratory Diseases Program, Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Division of Experimental Medicine, Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Salman T Qureshi
- Translational Research in Respiratory Diseases Program, Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Division of Experimental Medicine, Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
39
|
Na Pombejra S, Jamklang M, Uhrig JP, Vu K, Gelli A. The structure-function analysis of the Mpr1 metalloprotease determinants of activity during migration of fungal cells across the blood-brain barrier. PLoS One 2018; 13:e0203020. [PMID: 30161190 PMCID: PMC6117016 DOI: 10.1371/journal.pone.0203020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 08/14/2018] [Indexed: 12/21/2022] Open
Abstract
Cryptococcal meningoencephalitis, the most common form of cryptococcosis, is caused by the opportunistic fungal pathogen, Cryptococcus neoformans. Molecular strategies used by C. neoformans to invade the central nervous system (CNS) have been the focus of several studies. Recently, the role of a novel secreted metalloprotease (Mpr1) in the pathogenicity of C. neoformans was confirmed by studies demonstrating that Mpr1 mediated the migration of fungal cells into the CNS. Given this central function, the aim here was to identify the molecular determinants of Mpr1 activity and resolve their role in the migration of cryptococci across the blood-brain barrier (BBB). The Mpr1 protein belongs to an understudied group of metalloproteases of the M36 class of fungalysins unique to fungi. They are generally synthesized as propeptides with fairly long prodomains and highly conserved regions within their catalytic core. Through structure-function analysis of Mpr1, our study identified the prodomain cleavage sites of Mpr1 and demonstrated that when mutated, the prodomain appears to remain attached to the catalytic C-terminus of Mpr1 rendering a nonfunctional Mpr1 protein and an inability for cryptococci to cross the BBB. We found that proteolytic activity of Mpr1 was dependent on the coordination of zinc with two histidine residues in the active site of Mpr1, since amino acid substitutions in the HExxH motif abolished Mpr1 proteolytic activity and prevented the migration of cryptococci across the BBB. A phylogenetic analysis of Mpr1 revealed a distinct pattern likely reflecting the neurotropic nature of C. neoformans and the specific function of Mpr1 in breaching the BBB. This study contributes to a deeper understanding of the molecular regulation of Mpr1 activity and may lead to the development of specific inhibitors that could be used to restrict fungal penetration of the CNS and thus prevent cryptococcal meningoencephalitis-related deaths.
Collapse
Affiliation(s)
- Sarisa Na Pombejra
- Department of Pharmacology, School of Medicine, University of California, Davis, California, United States of America
| | - Mantana Jamklang
- Department of Pharmacology, School of Medicine, University of California, Davis, California, United States of America
| | - John P. Uhrig
- Department of Pharmacology, School of Medicine, University of California, Davis, California, United States of America
| | - Kiem Vu
- Department of Pharmacology, School of Medicine, University of California, Davis, California, United States of America
| | - Angie Gelli
- Department of Pharmacology, School of Medicine, University of California, Davis, California, United States of America
| |
Collapse
|
40
|
Casadevall A, Coelho C, Alanio A. Mechanisms of Cryptococcus neoformans-Mediated Host Damage. Front Immunol 2018; 9:855. [PMID: 29760698 PMCID: PMC5936990 DOI: 10.3389/fimmu.2018.00855] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 04/06/2018] [Indexed: 01/22/2023] Open
Abstract
Cryptococcus neoformans is not usually considered a cytotoxic fungal pathogen but there is considerable evidence that this microbe can damage host cells and tissues. In this essay, we review the evidence that C. neoformans damages host cells and note that the mechanisms involved are diverse. We consider C. neoformans-mediated host damage at the molecular, cellular, tissue, and organism level. Direct mechanisms of cytotoxicity include lytic exocytosis, organelle dysfunction, phagolysosomal membrane damage, and cytoskeletal alterations. Cytotoxicity contributes to pathogenesis by interfering with immune effector cell function and disrupting endothelial barriers thus allowing dissemination. When C. neoformans-mediated and immune-mediated host damage is sufficient to affect homeostasis, cryptococcosis occurs at the organism level.
Collapse
Affiliation(s)
- Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, United States
| | - Carolina Coelho
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, United States
| | - Alexandre Alanio
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, United States
- Institut Pasteur, Molecular Mycology Unit, CNRS UMR2000, Paris, France
- Laboratoire de Parasitologie-Mycologie, Hôpital Saint-Louis, Groupe Hospitalier Lariboisière, Saint-Louis, Fernand Widal, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
41
|
Esher SK, Zaragoza O, Alspaugh JA. Cryptococcal pathogenic mechanisms: a dangerous trip from the environment to the brain. Mem Inst Oswaldo Cruz 2018; 113:e180057. [PMID: 29668825 PMCID: PMC5909089 DOI: 10.1590/0074-02760180057] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 03/08/2018] [Indexed: 12/16/2022] Open
Abstract
Cryptococcus neoformans is an opportunistic pathogenic yeast that causes serious infections, most commonly of the central nervous system (CNS). C. neoformans is mainly found in the environment and acquired by inhalation. It could be metaphorically imagined that cryptococcal disease is a "journey" for the microorganism that starts in the environment, where this yeast loads its suitcase with virulence traits. C. neoformans first encounters the infected mammalian host in the lungs, a site in which it must choose the right elements from its "virulence suitcase" to survive the pulmonary immune response. However, the lung is often only the first stop in this journey, and in some individuals the fungal trip continues to the brain. To enter the brain, C. neoformans must "open" the main barrier that protects this organ, the blood brain barrier (BBB). Once in the brain, C. neoformans expresses a distinct set of protective attributes that confers a strong neurotropism and the ability to cause brain colonisation. In summary, C. neoformans is a unique fungal pathogen as shown in its ability to survive in the face of multiple stress factors and to express virulence factors that contribute to the development of disease.
Collapse
Affiliation(s)
- Shannon K Esher
- Duke University School of Medicine, Department of Molecular Genetics and Microbiology, Department of Medicine, Durham, USA
| | - Oscar Zaragoza
- Instituto de Salud Carlos III, National Centre for Microbiology, Mycology Reference Laboratory, Madrid, Spain
| | - James Andrew Alspaugh
- Duke University School of Medicine, Department of Molecular Genetics and Microbiology, Department of Medicine, Durham, USA
| |
Collapse
|
42
|
Aaron PA, Jamklang M, Uhrig JP, Gelli A. The blood-brain barrier internalises Cryptococcus neoformans via the EphA2-tyrosine kinase receptor. Cell Microbiol 2018; 20. [PMID: 29197141 DOI: 10.1111/cmi.12811] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 11/13/2017] [Accepted: 11/14/2017] [Indexed: 02/06/2023]
Abstract
Cryptococcus neoformans is an opportunistic fungal pathogen that causes life-threatening meningitis most commonly in populations with impaired immunity. Here, we resolved the transcriptome of the human brain endothelium challenged with C. neoformans to establish whether C. neoformans invades the CNS by co-opting particular signalling pathways as a means to promote its own entry. Among the 5 major pathways targeted by C. neoformans, the EPH-EphrinA1 (EphA2) tyrosine kinase receptor-signalling pathway was examined further. Silencing the EphA2 receptor transcript in a human brain endothelial cell line or blocking EphA2 activity with an antibody or chemical inhibitor prevented transmigration of C. neoformans in an in vitro model of the blood-brain barrier (BBB). In contrast, treating brain endothelial cells with an EphA2 chemical agonist or an EphA2 ligand promoted greater migration of fungal cells across the BBB. C. neoformans activated the EPH-tyrosine kinase pathway through a CD44-dependent phosphorylation of EphA2, promoting clustering and internalisation of EphA2 receptors. Moreover, HEK293T cells expressing EphA2 revealed an association between EphA2 and C. neoformans that boosted internalisation of C. neoformans. Collectively, the results suggest that C. neoformans promotes EphA2 activity via CD44, and this in turn creates a permeable barrier that facilitates the migration of C. neoformans across the BBB.
Collapse
Affiliation(s)
- Phylicia A Aaron
- Department of Pharmacology, School of Medicine, Genome and Biomedical Sciences Facility, University of California, Davis, California, USA
| | - Mantana Jamklang
- Department of Pharmacology, School of Medicine, Genome and Biomedical Sciences Facility, University of California, Davis, California, USA
| | - John P Uhrig
- Department of Pharmacology, School of Medicine, Genome and Biomedical Sciences Facility, University of California, Davis, California, USA
| | - Angie Gelli
- Department of Pharmacology, School of Medicine, Genome and Biomedical Sciences Facility, University of California, Davis, California, USA
| |
Collapse
|
43
|
Neuro-Immune Mechanisms of Anti-Cryptococcal Protection. J Fungi (Basel) 2017; 4:jof4010004. [PMID: 29371497 PMCID: PMC5872307 DOI: 10.3390/jof4010004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 12/10/2017] [Accepted: 12/23/2017] [Indexed: 12/15/2022] Open
Abstract
Cryptococcal meningitis (CM) is a life-threatening fungal disease affecting both immunosuppressed and immunocompetent people. The main causative agent of CM is Cryptococcus neoformans, a basidiomycete fungus prevalent in the environment. Our understanding of the immune mechanisms controlling C. neoformans growth within the central nervous system (CNS) is poor. However, there have been several recent advances in the field of neuroimmunology regarding how cells resident within the CNS, such as microglia and neurons, can participate in immune surveillance and control of infection. In this mini-review, the cells of the CNS are discussed with reference to what is currently known about how they control C. neoformans infection.
Collapse
|
44
|
Squizani ED, Oliveira NK, Reuwsaat JCV, Marques BM, Lopes W, Gerber AL, de Vasconcelos ATR, Lev S, Djordjevic JT, Schrank A, Vainstein MH, Staats CC, Kmetzsch L. Cryptococcal dissemination to the central nervous system requires the vacuolar calcium transporter Pmc1. Cell Microbiol 2017; 20. [PMID: 29113016 DOI: 10.1111/cmi.12803] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 10/27/2017] [Accepted: 10/30/2017] [Indexed: 12/26/2022]
Abstract
Cryptococcus neoformans is a basidiomycetous yeast and the cause of cryptococcosis in immunocompromised individuals. The most severe form of the disease is meningoencephalitis, which is one of the leading causes of death in HIV/AIDS patients. In order to access the central nervous system, C. neoformans relies on the activity of certain virulence factors such as urease, which allows transmigration through the blood-brain barrier. In this study, we demonstrate that the calcium transporter Pmc1 enables C. neoformans to penetrate the central nervous system, because the pmc1 null mutant failed to infect and to survive within the brain parenchyma in a murine systemic infection model. To investigate potential alterations in transmigration pathways in these mutants, global expression profiling of the pmc1 mutant strain was undertaken, and genes associated with urease, the Ca2+ -calcineurin pathway, and capsule assembly were identified as being differentially expressed. Also, a decrease in urease activity was observed in the calcium transporter null mutants. Finally, we demonstrate that the transcription factor Crz1 regulates urease activity and that the Ca2+ -calcineurin signalling pathway positively controls the transcription of calcium transporter genes and factors related to transmigration.
Collapse
Affiliation(s)
| | | | | | | | - William Lopes
- Centro de Biotecnologia, UFRGS, Porto Alegre, RS, Brazil
| | - Alexandra L Gerber
- Laboratório Nacional de Computação Científica (LNCC), Petrópolis, RJ, Brazil
| | | | - Sophie Lev
- Fungal Pathogenesis Laboratory, Centre for Infectious Diseases and Microbiology, The Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Julianne T Djordjevic
- Fungal Pathogenesis Laboratory, Centre for Infectious Diseases and Microbiology, The Westmead Institute for Medical Research, Westmead, NSW, Australia
| | | | | | | | - Lívia Kmetzsch
- Centro de Biotecnologia, UFRGS, Porto Alegre, RS, Brazil
| |
Collapse
|
45
|
Yang CL, Wang J, Zou LL. Innate immune evasion strategies against Cryptococcal meningitis caused by Cryptococcus neoformans. Exp Ther Med 2017; 14:5243-5250. [PMID: 29285049 PMCID: PMC5740712 DOI: 10.3892/etm.2017.5220] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 05/31/2017] [Indexed: 12/14/2022] Open
Abstract
As an infectious fungus that affects the respiratory tract, Cryptococcus neoformans (C. neoformans) commonly causes asymptomatic pulmonary infection. C. neoformans may target the brain instead of the lungs and cross the blood-brain barrier (BBB) in the early phase of infection; however, this is dependent on successful evasion of the host innate immune system. During the initial stage of fungal infection, a complex network of innate immune factors are activated. C. neoformans utilizes a number of strategies to overcome the anti-fungal mechanisms of the host innate immune system and cross the BBB. In the present review, the defensive mechanisms of C. neoformans against the innate immune system and its ability to cross the BBB were discussed, with an emphasis on recent insights into the activities of anti-phagocytotic and anti-oxidative factors in C. neoformans.
Collapse
Affiliation(s)
- Cheng-Liang Yang
- Translational Neuroscience and Neural Regeneration and Repair Institute, The First Hospital of Yichang, China Three Gorges University, Yichang, Hubei 443002, P.R. China.,Institute of Cell Therapy, The First Hospital of Yichang, China Three Gorges University, Yichang, Hubei 443002, P.R. China
| | - Jun Wang
- Translational Neuroscience and Neural Regeneration and Repair Institute, The First Hospital of Yichang, China Three Gorges University, Yichang, Hubei 443002, P.R. China.,Institute of Cell Therapy, The First Hospital of Yichang, China Three Gorges University, Yichang, Hubei 443002, P.R. China
| | - Li-Li Zou
- Translational Neuroscience and Neural Regeneration and Repair Institute, The First Hospital of Yichang, China Three Gorges University, Yichang, Hubei 443002, P.R. China.,Institute of Cell Therapy, The First Hospital of Yichang, China Three Gorges University, Yichang, Hubei 443002, P.R. China.,Department of Microbiology and Immunology, Medical College, China Three Gorges University, Yichang, Hubei 443002, P.R. China
| |
Collapse
|
46
|
Ruck T, Bittner S, Meuth SG, Herty M. Insights from mathematical modelling for T cell migration into the central nervous system. MATHEMATICAL MEDICINE AND BIOLOGY-A JOURNAL OF THE IMA 2017; 34:39-58. [PMID: 26519370 DOI: 10.1093/imammb/dqv038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Accepted: 10/08/2015] [Indexed: 01/23/2023]
Abstract
The migration of immune cells from peripheral immune organs into the central nervous system (CNS) through the blood-brain barrier (BBB) is a tightly regulated process. The complex interplay between cells of the BBB and immune cells coordinates cell migration as a part of normal immune surveillance while its dysregulation is critically involved in the pathogenesis of various CNS diseases. To develop tools for a deeper understanding of distribution and migratory pattern of immune cells regulated by the BBB, we made use of a mathematical modelling approach derived from Markov chain theory. We present a data-driven model using a derivation of kinetic differential equations from a particle game. According to the theory of gases, these equations allow one to predict the mean behaviour of a large class of cells by modelling cell-cell interactions. We used this model to assess the distribution of naive, central memory and effector memory T lymphocytes in the peripheral blood and cerebrospinal fluid. Our model allows us to evaluate the impact of activation status, migratory capacity and cell death for cell distribution in the peripheral blood and the CNS.
Collapse
|
47
|
Alves IA, Staudt KJ, Silva CDM, Lock GDA, Dalla Costa T, de Araujo BV. Influence of Experimental Cryptococcal Meningitis in Wistar Rats on Voriconazole Brain Penetration Assessed by Microdialysis. Antimicrob Agents Chemother 2017; 61:e00321-17. [PMID: 28483963 PMCID: PMC5487622 DOI: 10.1128/aac.00321-17] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 04/28/2017] [Indexed: 12/24/2022] Open
Abstract
To make advances in the treatment of cryptococcal meningitis, it is crucial to know a given drug's free fraction that reaches the biophase. In the present study, we applied microdialysis (μD) as a tool to determine the free levels reached by voriconazole (VRC) in the brains of healthy and Cryptococcus neoformans-infected rats. The infection was induced by the intravenous (i.v.) administration of 1 × 105 CFU of yeast. The dose administered was 5 mg/kg (of body weight) of VRC, given i.v. Plasma and microdialysate samples were analyzed by liquid chromatography-tandem mass spectrometry (LC-MS/MS) and LC-UV methods. The free brain/free plasma ratio (fT) and population pharmacokinetic (popPK) analyses were performed to evaluate the impact of infection on PK parameters of the drug. The brain penetration ratio showed an increase on brain exposure in infected animals (fThealthy = 0.85 versus fTinfected = 1.86). The structural PK model with two compartments and Michaelis-Menten (MM) elimination describes the VRC concentration-time profile in plasma and tissue simultaneously. The covariate infection was included in volume of distribution in the peripheral compartment in healthy animals (V2) and maximum rate of metabolism (VM ). The levels reached in infected tissues were higher than the values described for MIC of VRC for Cryptococccus neoformans (0.03 to 0.5 μg ml-1), indicating its great potential to treat meningitis associated with C. neoformans.
Collapse
Affiliation(s)
- Izabel Almeida Alves
- Pharmaceutical Sciences Graduate Program of Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Keli Jaqueline Staudt
- Medical Sciences Graduate Program of Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Carolina de Miranda Silva
- Pharmaceutical Sciences Graduate Program of Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Graziela de Araujo Lock
- Pharmaceutical Sciences Graduate Program of Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Teresa Dalla Costa
- Pharmaceutical Sciences Graduate Program of Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Bibiana Verlindo de Araujo
- Pharmaceutical Sciences Graduate Program of Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Medical Sciences Graduate Program of Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
48
|
Na Pombejra S, Salemi M, Phinney BS, Gelli A. The Metalloprotease, Mpr1, Engages AnnexinA2 to Promote the Transcytosis of Fungal Cells across the Blood-Brain Barrier. Front Cell Infect Microbiol 2017; 7:296. [PMID: 28713781 PMCID: PMC5492700 DOI: 10.3389/fcimb.2017.00296] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 06/16/2017] [Indexed: 12/19/2022] Open
Abstract
Eukaryotic pathogens display multiple mechanisms for breaching the blood-brain barrier (BBB) and invading the central nervous system (CNS). Of the fungal spp., that cause disease in mammals, only some cross brain microvascular endothelial cells which constitute the BBB, and invade the brain. Cryptococcus neoformans, the leading cause of fungal meningoencephalitis, crosses the BBB directly by transcytosis or by co-opting monocytes. We previously determined that Mpr1, a secreted fungal metalloprotease, facilitates association of fungal cells to brain microvascular endothelial cells and we confirmed that the sole expression of CnMPR1 endowed S. cerevisiae with an ability to cross the BBB. Here, the gain of function conferred onto S. cerevisiae by CnMPR1 (i.e., Sc<CnMPR1> strain) was used to identify targets of Mpr1 that might reside on the surface of the BBB. Following biotin-labeling of BBB surface proteins, Sc<CnMPR1>-associated proteins were identified by LC-MS/MS. Of the 62 proteins identified several were cytoskeleton-endocytosis-associated including AnnexinA2 (AnxA2). Using an in vitro model of the human BBB where AnxA2 activity was blocked, we found that the lack of AnxA2 activity prevented the movement of S. cerevisiae across the BBB (i.e., transcytosis of Sc<CnMPR1> strain) but unexpectedly, TEM analysis revealed that AnxA2 was not required for the association or the internalization of Sc<CnMPR1>. Additionally, the co-localization of AnxA2 and Sc<CnMPR1> suggest that successful crossing of the BBB is dependent on an AxnA2-Mpr1-mediated interaction. Collectively the data suggest that AnxA2 plays a central role in fungal transcytosis in human brain microvascular endothelial cells. The movement and exocytosis of Sc<CnMPR1> is dependent on membrane trafficking events that involve AnxA2 but these events appear to be independent from the actions of AnxA2 at the host cell surface. We propose that Mpr1 activity promotes cytoskeleton remodeling in brain microvascular endothelial cells and thereby engages AnxA2 in order to facilitate fungal transcytosis of the BBB.
Collapse
Affiliation(s)
- Sarisa Na Pombejra
- Department of Pharmacology, School of Medicine, Genome and Biomedical Sciences Facility, University of California, DavisDavis, CA, United States
| | - Michelle Salemi
- Proteomics Core Facility, Genome and Biomedical Sciences Facility, University of California, DavisDavis, CA, United States
| | - Brett S Phinney
- Proteomics Core Facility, Genome and Biomedical Sciences Facility, University of California, DavisDavis, CA, United States
| | - Angie Gelli
- Department of Pharmacology, School of Medicine, Genome and Biomedical Sciences Facility, University of California, DavisDavis, CA, United States
| |
Collapse
|
49
|
Hardy A, Benford D, Halldorsson T, Jeger MJ, Knutsen HK, More S, Naegeli H, Noteborn H, Ockleford C, Ricci A, Rychen G, Schlatter JR, Silano V, Solecki R, Turck D, Bresson JL, Dusemund B, Gundert-Remy U, Kersting M, Lambré C, Penninks A, Tritscher A, Waalkens-Berendsen I, Woutersen R, Arcella D, Court Marques D, Dorne JL, Kass GE, Mortensen A. Guidance on the risk assessment of substances present in food intended for infants below 16 weeks of age. EFSA J 2017; 15:e04849. [PMID: 32625502 PMCID: PMC7010120 DOI: 10.2903/j.efsa.2017.4849] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Following a request from the European Commission to EFSA, the EFSA Scientific Committee (SC) prepared a guidance for the risk assessment of substances present in food intended for infants below 16 weeks of age. In its approach to develop this guidance, the EFSA SC took into account, among others, (i) an exposure assessment based on infant formula as the only source of nutrition; (ii) knowledge of organ development in human infants, including the development of the gut, metabolic and excretory capacities, the brain and brain barriers, the immune system, the endocrine and reproductive systems; (iii) the overall toxicological profile of the substance identified through the standard toxicological tests, including critical effects; (iv) the relevance for the human infant of the neonatal experimental animal models used. The EFSA SC notes that during the period from birth up to 16 weeks, infants are expected to be exclusively fed on breast milk and/or infant formula. The EFSA SC views this period as the time where health-based guidance values for the general population do not apply without further considerations. High infant formula consumption per body weight is derived from 95th percentile consumption. The first weeks of life is the time of the highest relative consumption on a body weight basis. Therefore, when performing an exposure assessment, the EFSA SC proposes to use the high consumption value of 260 mL/kg bw per day. A decision tree approach is proposed that enables a risk assessment of substances present in food intended for infants below 16 weeks of age. The additional information needed when testing substances present in food for infants below 16 weeks of age and the approach to be taken for the risk assessment are on a case-by-case basis, depending on whether the substance is added intentionally to food and is systemically available.
Collapse
|
50
|
Trojan Horse Transit Contributes to Blood-Brain Barrier Crossing of a Eukaryotic Pathogen. mBio 2017; 8:mBio.02183-16. [PMID: 28143979 PMCID: PMC5285505 DOI: 10.1128/mbio.02183-16] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The blood-brain barrier (BBB) protects the central nervous system (CNS) by restricting the passage of molecules and microorganisms. Despite this barrier, however, the fungal pathogen Cryptococcus neoformans invades the brain, causing a meningoencephalitis that is estimated to kill over 600,000 people annually. Cryptococcal infection begins in the lung, and experimental evidence suggests that host phagocytes play a role in subsequent dissemination, although this role remains ill defined. Additionally, the disparate experimental approaches that have been used to probe various potential routes of BBB transit make it impossible to assess their relative contributions, confounding any integrated understanding of cryptococcal brain entry. Here we used an in vitro model BBB to show that a “Trojan horse” mechanism contributes significantly to fungal barrier crossing and that host factors regulate this process independently of free fungal transit. We also, for the first time, directly imaged C. neoformans-containing phagocytes crossing the BBB, showing that they do so via transendothelial pores. Finally, we found that Trojan horse crossing enables CNS entry of fungal mutants that cannot otherwise traverse the BBB, and we demonstrate additional intercellular interactions that may contribute to brain entry. Our work elucidates the mechanism of cryptococcal brain invasion and offers approaches to study other neuropathogens. The fungal pathogen Cryptococcus neoformans invades the brain, causing a meningoencephalitis that kills hundreds of thousands of people each year. One route that has been proposed for this brain entry is a Trojan horse mechanism, whereby the fungus crosses the blood-brain barrier (BBB) as a passenger inside host phagocytes. Although indirect experimental evidence supports this intriguing mechanism, it has never been directly visualized. Here we directly image Trojan horse transit and show that it is regulated independently of free fungal entry, contributes to cryptococcal BBB crossing, and allows mutant fungi that cannot enter alone to invade the brain.
Collapse
|