1
|
Oshika, Bari VK. Molecular mechanism of host-yeast interactions and prevention by nanoformulation approaches. Microb Pathog 2025; 205:107663. [PMID: 40339625 DOI: 10.1016/j.micpath.2025.107663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 04/17/2025] [Accepted: 05/02/2025] [Indexed: 05/10/2025]
Abstract
Fungal infections are a major source of morbidity and mortality in people with compromised immune systems, such as those with human immunodeficiency virus, cancer, organ transplant recipients, and patients undergoing chemotherapy in healthcare settings. According to a recent World Health Organization (WHO) fungal priority pathogens list, Cryptococcus spp., Candida spp., Aspergillus spp., and Candida auris cause severe invasive infections in human. These opportunistic pathogens cause a significant number of mycoses, which affect over a billion people annually. Around two million infections can be fatal, especially for those with compromised immune systems. To diagnose and treat mycoses, we need to understand the complex interactions between the fungus and the host during pathogenesis, the virulence-causing traits of the fungus, and how the host fights infection through the immune system. Although several antifungal drugs are available against fungal infections, their effectiveness is highly variable, with adverse effects. In addition, the increasing resistance to traditional antifungal treatments poses serious risks to the healthcare industry. Therefore, new therapeutic strategies are required to combat these potentially fatal fungal infections. Nanostructure-based formulations can improve the therapeutic efficacy of conventional medications by broadening their activities, decreasing toxicity, enhancing bioactivity, and improving biodistribution. The review highlights host and fungus interaction and how nanoformulations can be targeted against fungal infections.
Collapse
Affiliation(s)
- Oshika
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, VPO, Ghudda, Bathinda, India
| | - Vinay Kumar Bari
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, VPO, Ghudda, Bathinda, India.
| |
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
|
Al-Huthaifi AM, Radman BA, Al-Alawi AA, Mahmood F, Liu TB. Mechanisms and Virulence Factors of Cryptococcus neoformans Dissemination to the Central Nervous System. J Fungi (Basel) 2024; 10:586. [PMID: 39194911 DOI: 10.3390/jof10080586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 07/31/2024] [Accepted: 08/15/2024] [Indexed: 08/29/2024] Open
Abstract
Cryptococcosis is a prevalent fungal infection of the central nervous system (CNS) caused by Cryptococcus neoformans, a yeast with a polysaccharide capsule in the basidiomycete group. Normally, C. neoformans infects the respiratory tract and then breaches the blood-brain barrier (BBB), leading to meningitis or meningoencephalitis, which leads to hundreds of thousands of deaths each year. Although the mechanism by which C. neoformans infiltrates the BBB to invade the brain has yet to be fully understood, research has revealed that C. neoformans can cross the BBB using transcellular penetration, paracellular traversal, and infected phagocytes (the "Trojan horse" mechanism). The secretion of multiple virulence factors by C. neoformans is crucial in facilitating the spread of infection after breaching the BBB and causing brain infections. Extensive research has shown that various virulence factors play a significant role in the dissemination of infection beyond the lungs. This review explores the mechanisms of C. neoformans entering the CNS and explains how it bypasses the BBB. Additionally, it aims to understand the interplay between the regulatory mechanisms and virulence factors of C. neoformans.
Collapse
Affiliation(s)
| | - Bakeel A Radman
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
| | | | - Fawad Mahmood
- Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Tong-Bao Liu
- Medical Research Institute, Southwest University, Chongqing 400715, China
- Jinfeng Laboratory, Chongqing 401329, China
- Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing 400715, China
| |
Collapse
|
4
|
Davis JM. A deep cut into early cryptococcal pathogenesis. mBio 2024; 15:e0065724. [PMID: 38975784 PMCID: PMC11323497 DOI: 10.1128/mbio.00657-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2024] Open
Abstract
Dissemination from one organ system to another is common to many pathogens and often the key process separating simple illness from fatal infection. The pathogenic Cryptococcus species offer a prime example. Cryptococcal infection is thought to begin in the lungs, as a mild or asymptomatic pneumonia. However, bloodborne dissemination from the lungs to the brain is responsible for the most devastating forms of infection. As with other disseminating infections, the transition likely depends on rare but crucial events, such as the crossing of a tissue barrier. By their nature, these events are difficult to study. Francis et al. (mBio 15:e03078-23, 2024, https://doi.org/10.1128/mbio.03078-23) have addressed this difficulty by developing a powerful imaging pipeline to scan through unprecedented volumes of tissue from mice infected with Cryptococcus at multiple stages of infection. Their observations challenge some of our basic assumptions about cryptococcal pathogenesis, including when and how the organism reaches the bloodstream and the central nervous system.
Collapse
Affiliation(s)
- J. Muse Davis
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
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
|
8
|
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
|
9
|
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
|
10
|
Sun S, Roth C, Floyd Averette A, Magwene PM, Heitman J. Epistatic genetic interactions govern morphogenesis during sexual reproduction and infection in a global human fungal pathogen. Proc Natl Acad Sci U S A 2022; 119:e2122293119. [PMID: 35169080 PMCID: PMC8872808 DOI: 10.1073/pnas.2122293119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/11/2022] [Indexed: 12/12/2022] Open
Abstract
Cellular development is orchestrated by evolutionarily conserved signaling pathways, which are often pleiotropic and involve intra- and interpathway epistatic interactions that form intricate, complex regulatory networks. Cryptococcus species are a group of closely related human fungal pathogens that grow as yeasts yet transition to hyphae during sexual reproduction. Additionally, during infection they can form large, polyploid titan cells that evade immunity and develop drug resistance. Multiple known signaling pathways regulate cellular development, yet how these are coordinated and interact with genetic variation is less well understood. Here, we conducted quantitative trait locus (QTL) analyses of a mapping population generated by sexual reproduction of two parents, only one of which is unisexually fertile. We observed transgressive segregation of the unisexual phenotype among progeny, as well as a large-cell phenotype under mating-inducing conditions. These large-cell progeny were found to produce titan cells both in vitro and in infected animals. Two major QTLs and corresponding quantitative trait genes (QTGs) were identified: RIC8 (encoding a guanine-exchange factor) and CNC06490 (encoding a putative Rho-GTPase activator), both involved in G protein signaling. The two QTGs interact epistatically with each other and with the mating-type locus in phenotypic determination. These findings provide insights into the complex genetics of morphogenesis during unisexual reproduction and pathogenic titan cell formation and illustrate how QTL analysis can be applied to identify epistasis between genes. This study shows that phenotypic outcomes are influenced by the genetic background upon which mutations arise, implicating dynamic, complex genotype-to-phenotype landscapes in fungal pathogens and beyond.
Collapse
Affiliation(s)
- Sheng Sun
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710
| | - Cullen Roth
- Department of Biology, Duke University, Durham, NC 27708
| | - Anna Floyd Averette
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710
| | - Paul M Magwene
- Department of Biology, Duke University, Durham, NC 27708
| | - Joseph Heitman
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710;
| |
Collapse
|
11
|
Souza TN, Valdez AF, Rizzo J, Zamith-Miranda D, Guimarães AJ, Nosanchuk JD, Nimrichter L. Host cell membrane microdomains and fungal infection. Cell Microbiol 2021; 23:e13385. [PMID: 34392593 PMCID: PMC8664998 DOI: 10.1111/cmi.13385] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/14/2021] [Accepted: 07/24/2021] [Indexed: 01/13/2023]
Abstract
Lipid microdomains or lipid rafts are dynamic and tightly ordered regions of the plasma membrane. In mammalian cells, they are enriched in cholesterol, glycosphingolipids, Glycosylphosphatidylinositol-anchored and signalling-related proteins. Several studies have suggested that mammalian pattern recognition receptors are concentrated or recruited to lipid domains during host-pathogen association to enhance the effectiveness of host effector processes. However, pathogens have also evolved strategies to exploit these domains to invade cells and survive. In fungal organisms, a complex cell wall network usually mediates the first contact with the host cells. This cell wall may contain virulence factors that interfere with the host membrane microdomains dynamics, potentially impacting the infection outcome. Indeed, the microdomain disruption can dampen fungus-host cell adhesion, phagocytosis and cellular immune responses. Here, we provide an overview of regulatory strategies employed by pathogenic fungi to engage with and potentially subvert the lipid microdomains of host cells. TAKE AWAY: Lipid microdomains are ordered regions of the plasma membrane enriched in cholesterol, glycosphingolipids (GSL), GPI-anchored and signalling-related proteins. Pathogen recognition by host immune cells can involve lipid microdomain participation. During this process, these domains can coalesce in larger complexes recruiting receptors and signalling proteins, significantly increasing their signalling abilities. The antifungal innate immune response is mediated by the engagement of pathogen-associated molecular patterns to pattern recognition receptors (PRRs) at the plasma membrane of innate immune cells. Lipid microdomains can concentrate or recruit PRRs during host cell-fungi association through a multi-interactive mechanism. This association can enhance the effectiveness of host effector processes. However, virulence factors at the fungal cell surface and extracellular vesicles can re-assembly these domains, compromising the downstream signalling and favouring the disease development. Lipid microdomains are therefore very attractive targets for novel drugs to combat fungal infections.
Collapse
Affiliation(s)
- Taiane N Souza
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alessandro F Valdez
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Juliana Rizzo
- Unité Biologie des ARN des Pathogènes Fongiques, Département de Mycologie, Institut Pasteur, Paris, France
| | - Daniel Zamith-Miranda
- Departments of Medicine (Division of Infectious Diseases) and Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Allan Jefferson Guimarães
- Departamento de Microbiologia e Parasitologia-MIP, Instituto Biomédico, Universidade Federal Fluminense, Rio de Janeiro, Brazil
| | - Joshua D Nosanchuk
- Departments of Medicine (Division of Infectious Diseases) and Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Leonardo Nimrichter
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
12
|
Gao L, Zhao R, Liu J, Zhang W, Sun F, Yin Q, Wang X, Wang M, Feng T, Qin Y, Cai W, Li Q, Dong H, Chen X, Xiong X, Liu H, Hu J, Chen W, Han B. KIF15 Promotes Progression of Castration Resistant Prostate Cancer by Activating EGFR Signaling Pathway. Front Oncol 2021; 11:679173. [PMID: 34804913 PMCID: PMC8599584 DOI: 10.3389/fonc.2021.679173] [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] [Received: 03/11/2021] [Accepted: 10/18/2021] [Indexed: 11/29/2022] Open
Abstract
Castration-resistant prostate cancer (CRPC) continues to be a major clinical problem and its underlying mechanisms are still not fully understood. The epidermal growth factor receptor (EGFR) activation is an important event that regulates mitogenic signaling. EGFR signaling plays an important role in the transition from androgen dependence to castration-resistant state in prostate cancer (PCa). Kinesin family member 15 (KIF15) has been suggested to be overexpressed in multiple malignancies. Here, we demonstrate that KIF15 expression is elevated in CRPC. We show that KIF15 contributes to CRPC progression by enhancing the EGFR signaling pathway, which includes complex network intermediates such as mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3K)/AKT pathways. In CRPC tumors, increased expression of KIF15 is positively correlated with EGFR protein level. KIF15 binds to EGFR, and prevents EGFR proteins from degradation in a Cdc42-dependent manner. These findings highlight the key role of KIF15 in the development of CRPC and rationalize KIF15 as a potential therapeutic target.
Collapse
Affiliation(s)
- Lin Gao
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ru Zhao
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Junmei Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wenbo Zhang
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Feifei Sun
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qianshuo Yin
- School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xin Wang
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Meng Wang
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tingting Feng
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yiming Qin
- College of Chemical Engineering and Materials Science, Shandong Normal University, Jinan, China
| | - Wenjie Cai
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qianni Li
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hanchen Dong
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xueqing Chen
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xueting Xiong
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Hui Liu
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Pathology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jing Hu
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Pathology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Weiwen Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bo Han
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Pathology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
13
|
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
|
14
|
Gaylord EA, Choy HL, Doering TL. Dangerous Liaisons: Interactions of Cryptococcus neoformans with Host Phagocytes. Pathogens 2020; 9:E891. [PMID: 33121050 PMCID: PMC7692806 DOI: 10.3390/pathogens9110891] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 10/22/2020] [Accepted: 10/25/2020] [Indexed: 02/07/2023] Open
Abstract
Cryptococcus neoformans is an opportunistic fungal pathogen and a leading cause of death in immunocompromised individuals. The interactions of this yeast with host phagocytes are critical to disease outcome, and C. neoformans is equipped with an array of factors to modulate these processes. Cryptococcal infection begins with the deposition of infectious particles into the lungs, where the fungal cells deploy various antiphagocytic factors to resist internalization by host cells. If the cryptococci are still engulfed, they can survive and proliferate within host cells by modulating the phagolysosome environment in which they reside. Lastly, cryptococcal cells may escape from phagocytes by host cell lysis, nonlytic exocytosis, or lateral cell-to-cell transfer. The interactions between C. neoformans and host phagocytes also influence the dissemination of this pathogen to the brain, where it may cross the blood-brain barrier and cause an often-fatal meningoencephalitis. In this review, we highlight key cryptococcal factors involved in various stages of cryptococcal-host interaction and pathogenesis.
Collapse
Affiliation(s)
| | | | - Tamara L. Doering
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110, USA; (E.A.G.); (H.L.C.)
| |
Collapse
|
15
|
Ma Z, Peng J, Yu D, Park JS, Lin H, Xu B, Lu C, Fan H, Waldor MK. A streptococcal Fic domain-containing protein disrupts blood-brain barrier integrity by activating moesin in endothelial cells. PLoS Pathog 2019; 15:e1007737. [PMID: 31071198 PMCID: PMC6529018 DOI: 10.1371/journal.ppat.1007737] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 05/21/2019] [Accepted: 03/27/2019] [Indexed: 01/04/2023] Open
Abstract
Streptococcus equi subsp. zooepidemicus (SEZ) is a zoonotic pathogen capable of causing meningitis in humans. The mechanisms that enable pathogens to traverse the blood-brain barrier (BBB) are incompletely understood. Here, we investigated the role of a newly identified Fic domain-containing protein, BifA, in SEZ virulence. BifA was required for SEZ to cross the BBB and to cause meningitis in mice. BifA also enhanced SEZ translocation across human Brain Microvascular Endothelial Cell (hBMEC) monolayers. Purified BifA or its Fic domain-containing C-terminus alone were able to enter into hBMECs, leading to disruption of monolayer barrier integrity. A SILAC-based proteomic screen revealed that BifA binds moesin. BifA’s Fic domain was required for its binding to this regulator of host cell cytoskeletal processes. BifA treatment of hBMECs led to moesin phosphorylation and downstream RhoA activation. Inhibition of moesin activation or moesin depletion in hBMEC monolayers abrogated BifA-mediated increases in barrier permeability and SEZ’s capacity to translocate across monolayers. Thus, BifA activation of moesin appears to constitute a key mechanism by which SEZ disrupts endothelial monolayer integrity to penetrate the BBB. Streptococcus equi subsp. zooepidemicus (SEZ) is an important animal pathogen and can cause meningitis in humans. Little is known about how this Group C streptococcal species penetrates the blood-brain barrier (BBB). We identified bifA, a gene that is critical for SEZ to cause meningitis in mice and to penetrate a human brain endothelial monolayer in a tissue culture model. BifA’s Fic domain enables the protein to enter into endothelial monolayers and to bind to moesin, a cytoskeletal regulatory protein, leading to its activation. Preventing moesin activation abolished BifA-induced barrier leakiness and SEZ’s capacity to penetrate a monolayer barrier. Together, our findings suggest that SEZ meningitis depends on BifA, a Fic-domain protein that manipulates moesin-dependent signaling to modulate BBB permeability.
Collapse
Affiliation(s)
- Zhe Ma
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
- Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, United States of America
- Ministry of Agriculture Key Laboratory of Animal Bacteriology, Nanjing, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China
| | - Jie Peng
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Dandan Yu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Joseph S. Park
- Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, United States of America
- Howard Hughes Medical Institute, Boston, Massachusetts, United States of America
| | - Huixing Lin
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
- Ministry of Agriculture Key Laboratory of Animal Bacteriology, Nanjing, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China
| | - Bin Xu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Chengping Lu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Hongjie Fan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
- Ministry of Agriculture Key Laboratory of Animal Bacteriology, Nanjing, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China
- * E-mail: (HF); (MKW)
| | - Matthew K. Waldor
- Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, United States of America
- Howard Hughes Medical Institute, Boston, Massachusetts, United States of America
- * E-mail: (HF); (MKW)
| |
Collapse
|
16
|
Min P, Zhao S, Liu L, Zhang Y, Ma Y, Zhao X, Wang Y, Song Y, Zhu C, Jiang H, Gu L, Du J. MICAL-L2 potentiates Cdc42-dependent EGFR stability and promotes gastric cancer cell migration. J Cell Mol Med 2019; 23:4475-4488. [PMID: 31034158 PMCID: PMC6533512 DOI: 10.1111/jcmm.14353] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 03/29/2019] [Accepted: 04/12/2019] [Indexed: 01/21/2023] Open
Abstract
Enhanced migration potential is a common characteristic of cancer cells induced by mechanisms that are incompletely defined. The present study was designed to investigate relationship of a new discovered cytoskeleton regulator MICAL‐L2 and the endogenous epidermal growth factor receptor (EGFR) signalling pathways in gastric cancer cell migration. Increased expression of MICAL‐L2 in gastric cancer cells up‐regulated EGFR protein level, accompanied by the increase of cell migration, whereas silencing MICAL‐L2 down‐regulated EGFR and inhibited cell migration. Expression of MICAL‐L2 was also shown positively correlated with the activation of HSP27/cytoskeleton and HSP27/β‐catenin signalling pathways that provide key mechanisms controlling cell migration. The up‐regulating effect of MICAL‐L2 on EGFR is mediated through a transcription‐independent mechanism that involves inhibiting EGFR protein degradation in lysosome. Further analysis indicated that Cdc42 activation contributed in maintaining the effect of MICAL‐L2 on EGFR stability. Furthermore analysis of clinic specimens revealed increased expression of MICAL‐L2 in carcinoma tissues and a positive correlation between MICAL‐L2 and EGFR expression levels. The above results indicate that MICAL‐L2 potentiates gastric cell migration via inhibiting EGFR degradation in lysosome via a Cdc42‐dependent manner that leads to the activation of EGFR/HSP27 signalling pathways.
Collapse
Affiliation(s)
- Pengxiang Min
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shuo Zhao
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lei Liu
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.,Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yujie Zhang
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yadong Ma
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xuyang Zhao
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.,Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yueyuan Wang
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yixuan Song
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chenchen Zhu
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Haonan Jiang
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Luo Gu
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.,Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jun Du
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
17
|
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
|
18
|
Camacho E, Casadevall A. Cryptococcal Traits Mediating Adherence to Biotic and Abiotic Surfaces. J Fungi (Basel) 2018; 4:jof4030088. [PMID: 30060601 PMCID: PMC6162697 DOI: 10.3390/jof4030088] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 07/25/2018] [Accepted: 07/27/2018] [Indexed: 01/22/2023] Open
Abstract
Several species in the genus Cryptococcus are facultative intracellular pathogens capable of causing disease associated with high mortality and morbidity in humans. These fungi interact with other organisms in the soil, and these interactions may contribute to the development of adaptation mechanisms that function in virulence by promoting fungal survival in animal hosts. Fungal adhesion molecules, also known as adhesins, have been classically considered as cell-surface or secreted proteins that play critical roles in microbial pathogenesis or in biofilm formation as structural components. Pathogenic Cryptococcus spp. differ from other pathogenic yeasts in having a polysaccharide capsule that covers the cell wall surface and precludes interactions of those structures with host cell receptors. Hence, pathogenic Cryptococcus spp. use unconventional tools for surface attachment. In this essay, we review the unique traits and mechanisms favoring adhesion of Cryptococcus spp. to biotic and abiotic surfaces. Knowledge of the traits that mediate adherence could be exploited in the development of therapeutic, biomedical, and/or industrial products.
Collapse
Affiliation(s)
- Emma Camacho
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, 615 N Wolfe St Room E5132, Baltimore, MD 21205, USA.
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, 615 N Wolfe St Room E5132, Baltimore, MD 21205, USA.
| |
Collapse
|
19
|
Li M, Wen Y, Zhang R, Xie F, Zhang G, Qin X. Adenoviral vector-induced silencing of RGMa attenuates blood-brain barrier dysfunction in a rat model of MCAO/reperfusion. Brain Res Bull 2018; 142:54-62. [PMID: 29935233 DOI: 10.1016/j.brainresbull.2018.06.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 06/15/2018] [Accepted: 06/18/2018] [Indexed: 12/29/2022]
Abstract
BACKGROUND Repulsive guidance molecule A (RGMa) is implicated in focal cerebral ischemia-reperfusion (I/R) injury, but its mechanisms are still largely unknown. This work focused on the effects of RGMa on the blood-brain barrier (BBB) after focal cerebral I/R injury. METHODS Sprague-Dawley (SD) rats were randomly divided into four groups: sham, middle cerebral artery occlusion (MCAO)/reperfusion (I/R), MCAO/reperfusion administered recombinant adenovirus expressing sh-con (I/R + sh-con) and MCAO/reperfusion administered recombinant adenovirus expressing sh-RGMa (I/R + sh-RGMa) groups. Infarct volume, brain edema and neurological scores were evaluated at 3 day after reperfusion. Evens blue leakage and transmission electron microscopy was performed. And the expression level of claudin-5 and ZO-1, CDC-42 and PAK-1, RGMa were detected by western blot. RESULTS Compared with I/R or I/R + sh-con groups, I/R + sh-RGMa group showed smaller infarction volume, attenuated brain edema, improved neurological scores and better BBB integrity, such as reduced Evans blue leakage and ultra-structural change. We also observed improved BBB function followed by down-regulation of MMP-9 and up-regulation of claudin-5 and ZO-1 in the I/R + sh-RGMa group. In addition, up-regulation of the CDC-42 and PAK-1 in the I/R + sh-RGMa group was obtained. CONCLUSIONS RGMa may be involved in I/R injury associated with BBB dysfunction via the CDC-42/PAK-1 signal pathway and may be a promising therapeutic target for I/R injury.
Collapse
Affiliation(s)
- Min Li
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Neurology, Inner Mongolia People's Hospital, Hohhot, China
| | - Yuetao Wen
- Department of Neurosurgery, University-Town Hospital of Chongqing Medical University, Chongqing, China
| | - Rongrong Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fei Xie
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Gang Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xinyue Qin
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
20
|
Liu H, Jia L, Guo W, Sun Y, Zhu R, Li S, Qu G, Jiang H, Wang J, Gu J, Sun C, Feng X, Han W, Lei L. Differential Protein Profiling of Cerebrospinal Fluid in Piglets with Severe Meningoencephalitis Caused by Streptococcus suis Type 2 Compared to Controls. Front Cell Infect Microbiol 2018; 8:35. [PMID: 29479521 PMCID: PMC5811643 DOI: 10.3389/fcimb.2018.00035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 01/25/2018] [Indexed: 11/19/2022] Open
Abstract
Streptococcus suis serotype 2 (SS2) is a zoonotic pathogen that can cause meningitis both in pigs and in human beings. However, the pathogenesis of central nervous system (CNS) infection caused by SS2 have not yet been elucidated. To find the key molecules in cerebrospinal fluid (CSF) needed for the pathogenesis, a SS2 meningoencephalitic pig model and a SS2 non-meningoencephalitic pig model were established in this study. CSF was collected from infected piglets, and protein profiling was performed with label-free proteomics technology. A total of 813 differential proteins, including 52 up-regulated proteins and 761 down-regulated proteins, were found in the CSF of meningoencephalitic pigs compared with both non-meningoencephalitic pigs and healthy pigs. These 813 differential proteins were clustered into three main categories, namely, cellular component, biological process, and molecular function by gene ontology (GO) analysis. The most enriched subclasses of differential proteins in each category were exosome (44.3%), energy pathway (25.0%) and catalytic activity (11.3%), respectively. The most enriched subclasses of upregulated proteins were extracellular (62.1%), protein metabolism (34.5%) and cysteine-type peptidase activity (6.9%), and of downregulated proteins were exosomes (45.0%), energy pathway (24.0%) and catalytic activity (9.4%). Then, the differential proteins were further investigated by using the KEGG database and were found to participate in 16 KEGGs. The most enriched KEGG was citrate cycle (56.6%), and some of these differential proteins are associated with brain diseases such as Huntington's disease (18.6%), Parkinson's disease (23.8%) and Alzheimer's disease (17.6%). Sixteen of the 813 differential proteins, chosen randomly as examples, were further confirmed by enzyme-linked immunosorbent assay (ELISA) to support the proteomic data. To our knowledge, this is the first study to analyze the differential protein profiling of CSF between SS2 meningoencephalitic piglets and non-meningoencephalitic piglets by employing proteomic technology. The discovery and bioinformatics analysis of these differential proteins provides reference data not only for research on pathogenesis of SS2 CNS infection but also for diagnosis and drug therapy research.
Collapse
Affiliation(s)
- Hongtao Liu
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Li Jia
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Wenfei Guo
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yingying Sun
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Rining Zhu
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Shuguang Li
- Shandong Binzhou Animal Science and Veterinary Medicine Academy, Binzhou, China
| | - Guanggang Qu
- Shandong Binzhou Animal Science and Veterinary Medicine Academy, Binzhou, China
| | - Hexiang Jiang
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Junjie Wang
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jingmin Gu
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Changjiang Sun
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xin Feng
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Wenyu Han
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Liancheng Lei
- College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
21
|
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: 49] [Impact Index Per Article: 7.0] [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
|
22
|
Pandey A, Ding SL, Qin QM, Gupta R, Gomez G, Lin F, Feng X, Fachini da Costa L, Chaki SP, Katepalli M, Case ED, van Schaik EJ, Sidiq T, Khalaf O, Arenas A, Kobayashi KS, Samuel JE, Rivera GM, Alaniz RC, Sze SH, Qian X, Brown WJ, Rice-Ficht A, Russell WK, Ficht TA, de Figueiredo P. Global Reprogramming of Host Kinase Signaling in Response to Fungal Infection. Cell Host Microbe 2017; 21:637-649.e6. [PMID: 28494245 DOI: 10.1016/j.chom.2017.04.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 03/12/2017] [Accepted: 04/24/2017] [Indexed: 12/26/2022]
Abstract
Cryptococcus neoformans (Cn) is a deadly fungal pathogen whose intracellular lifestyle is important for virulence. Host mechanisms controlling fungal phagocytosis and replication remain obscure. Here, we perform a global phosphoproteomic analysis of the host response to Cryptococcus infection. Our analysis reveals numerous and diverse host proteins that are differentially phosphorylated following fungal ingestion by macrophages, thereby indicating global reprogramming of host kinase signaling. Notably, phagocytosis of the pathogen activates the host autophagy initiation complex (AIC) and the upstream regulatory components LKB1 and AMPKα, which regulate autophagy induction through their kinase activities. Deletion of Prkaa1, the gene encoding AMPKα1, in monocytes results in resistance to fungal colonization of mice. Finally, the recruitment of AIC components to nascent Cryptococcus-containing vacuoles (CnCVs) regulates the intracellular trafficking and replication of the pathogen. These findings demonstrate that host AIC regulatory networks confer susceptibility to infection and establish a proteomic resource for elucidating host mechanisms that regulate fungal intracellular parasitism.
Collapse
Affiliation(s)
- Aseem Pandey
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, College Station, Texas 77843, USA; Norman Borlaug Center, Texas A&M University, College Station, Texas 77843, USA; Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, Texas 77843, USA
| | - Sheng Li Ding
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, College Station, Texas 77843, USA; Norman Borlaug Center, Texas A&M University, College Station, Texas 77843, USA; Department of Plant Pathology, College of Plant Protection, Henan Agricultural University, Zhengzhou 450002, Henan, China
| | - Qing-Ming Qin
- College of Plant Sciences, Jilin University, Changchun 130062, Jilin, China; Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun 130062, Jilin, China
| | - Rahul Gupta
- Health and Engineering Group, Leidos Inc., 2295 Parklake Drive, Atlanta, GA 30345, USA
| | - Gabriel Gomez
- Texas A&M Veterinary Medical Diagnostic Laboratory, Texas A&M University, College Station, Texas 77843, USA
| | - Furong Lin
- Norman Borlaug Center, Texas A&M University, College Station, Texas 77843, USA
| | - Xuehuan Feng
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, College Station, Texas 77843, USA; Norman Borlaug Center, Texas A&M University, College Station, Texas 77843, USA
| | - Luciana Fachini da Costa
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, College Station, Texas 77843, USA; Norman Borlaug Center, Texas A&M University, College Station, Texas 77843, USA; Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, Texas 77843, USA
| | - Sankar P Chaki
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, Texas 77843, USA
| | - Madhu Katepalli
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, College Station, Texas 77843, USA
| | - Elizabeth D Case
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, College Station, Texas 77843, USA
| | - Erin J van Schaik
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, College Station, Texas 77843, USA
| | - Tabasum Sidiq
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, College Station, Texas 77843, USA
| | - Omar Khalaf
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, Texas 77843, USA
| | - Angela Arenas
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, Texas 77843, USA
| | - Koichi S Kobayashi
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, College Station, Texas 77843, USA
| | - James E Samuel
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, College Station, Texas 77843, USA
| | - Gonzalo M Rivera
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, Texas 77843, USA
| | - Robert C Alaniz
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, College Station, Texas 77843, USA
| | - Sing-Hoi Sze
- Center for Bioinformatics & Genomic Systems Engineering, Texas A&M University, College Station, Texas 77843, USA; Department of Computer Science and Engineering, Dwight Look College of Engineering, Texas A&M University, College Station, Texas 77843, USA; Department of Biochemistry & Biophysics, Texas A&M University, College Station, Texas 77843, USA
| | - Xiaoning Qian
- Center for Bioinformatics & Genomic Systems Engineering, Texas A&M University, College Station, Texas 77843, USA; Department of Electrical and Computer Engineering, Dwight Look College of Engineering, Texas A&M University, College Station, Texas 77843, USA
| | - William J Brown
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853-2703, USA
| | - Allison Rice-Ficht
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M Health Science Center, College Station, Texas 77843, USA
| | - William K Russell
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Thomas A Ficht
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, Texas 77843, USA.
| | - Paul de Figueiredo
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, College Station, Texas 77843, USA; Norman Borlaug Center, Texas A&M University, College Station, Texas 77843, USA; Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, Texas 77843, USA.
| |
Collapse
|
23
|
Heinisch JJ, Rodicio R. Protein kinase C in fungi—more than just cell wall integrity. FEMS Microbiol Rev 2017; 42:4562651. [DOI: 10.1093/femsre/fux051] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 10/19/2017] [Indexed: 11/13/2022] Open
|
24
|
Jamerson M, Schmoyer JA, Park J, Marciano-Cabral F, Cabral GA. Identification of Naegleria fowleri proteins linked to primary amoebic meningoencephalitis. MICROBIOLOGY-SGM 2017; 163:322-332. [PMID: 28086072 DOI: 10.1099/mic.0.000428] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Naegleria fowleri (N. fowleri) causes primary amoebic meningoencephalitis, a rapidly fatal disease of the central nervous system. N. fowleri can exist in cyst, flagellate or amoebic forms, depending on environmental conditions. The amoebic form can invade the brain following introduction into the nasal passages. When applied intranasally to a mouse model, cultured N. fowleri amoebae exhibit low virulence. However, upon serial passage in mouse brain, the amoebae acquire a highly virulent state. In the present study, a proteomics approach was applied to the identification of N. fowleri amoeba proteins whose expression was associated with the highly virulent state in mice. Mice were inoculated intranasally with axenically cultured amoebae or with mouse-passaged amoebae. Examination by light and electron microscopy revealed no morphological differences. However, mouse-passaged amoebae were more virulent in mice as indicated by exhibiting a two log10 titre decrease in median infective dose 50 (ID50). Scatter plot analysis of amoebic lysates revealed a subset of proteins, the expression of which was associated with highly virulent amoebae. MS-MS indicated that this subset contained proteins that shared homology with those linked to cytoskeletal rearrangement and the invasion process. Invasion assays were performed in the presence of a select inhibitor to expand on the findings. The collective results suggest that N. fowleri gene products linked to cytoskeletal rearrangement and invasion may be candidate targets in the management of primary amoebic meningoencephalitis.
Collapse
Affiliation(s)
- Melissa Jamerson
- Department of Clinical Laboratory Sciences, Virginia Commonwealth University School of Allied Health Professions, Richmond, VA 23298-0583, USA.,Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298-0678, USA
| | - Jacqueline A Schmoyer
- Department of Clinical Laboratory Sciences, Virginia Commonwealth University School of Allied Health Professions, Richmond, VA 23298-0583, USA
| | - Jay Park
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298-0678, USA
| | - Francine Marciano-Cabral
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298-0678, USA
| | - Guy A Cabral
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298-0678, USA
| |
Collapse
|
25
|
Selvan LDN, Sreenivasamurthy SK, Kumar S, Yelamanchi SD, Madugundu AK, Anil AK, Renuse S, Nair BG, Gowda H, Mathur PP, Satishchandra P, Shankar SK, Mahadevan A, Keshava Prasad TS. Characterization of host response to Cryptococcus neoformans through quantitative proteomic analysis of cryptococcal meningitis co-infected with HIV. MOLECULAR BIOSYSTEMS 2016; 11:2529-40. [PMID: 26181685 DOI: 10.1039/c5mb00187k] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Cryptococcal meningitis is the most common opportunistic fungal infection causing morbidity and mortality (>60%) in HIV-associated immunocompromised individuals caused by Cryptococcus neoformans. Molecular mechanisms of cryptococcal infection in brain have been studied using experimental animal models and cell lines. There are limited studies for the molecular understanding of cryptococcal meningitis in human brain. The proteins involved in the process of invasion and infection in human brain still remains obscure. To this end we carried out mass spectrometry-based quantitative proteomics of frontal lobe brain tissues from cryptococcal meningitis patients and controls to identify host proteins that are associated with the pathogenesis of cryptococcal meningitis. We identified 317 proteins to be differentially expressed (≥2-fold) from a total of 3423 human proteins. We found proteins involved in immune response and signal transduction to be differentially expressed in response to cryptococcal infection in human brain. Immune response proteins including complement factors, major histocompatibility proteins, proteins previously known to be involved in fungal invasion to brain such as caveolin 1 and actin were identified to be differentially expressed in cryptococcal meningitis brain tissues co-infected with HIV. We also validated the expression status of 5 proteins using immunohistochemistry. Overexpression of major histocompatibility complexes, class I, B (HLA-B), actin alpha 2 smooth muscle aorta (ACTA2) and caveolin 1 (CAV1) and downregulation of peripheral myelin protein 2 (PMP2) and alpha crystallin B chain (CRYAB) in cryptococcal meningitis were confirmed by IHC-based validation experiments. This study provides the brain proteome profile of cryptococcal meningitis co-infected with HIV for a better understanding of the host response associated with the disease.
Collapse
|
26
|
Ferreira ÉR, Horjales E, Bonfim-Melo A, Cortez C, da Silva CV, De Groote M, Sobreira TJP, Cruz MC, Lima FM, Cordero EM, Yoshida N, da Silveira JF, Mortara RA, Bahia D. Unique behavior of Trypanosoma cruzi mevalonate kinase: A conserved glycosomal enzyme involved in host cell invasion and signaling. Sci Rep 2016; 6:24610. [PMID: 27113535 PMCID: PMC4845012 DOI: 10.1038/srep24610] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 03/29/2016] [Indexed: 11/30/2022] Open
Abstract
Mevalonate kinase (MVK) is an essential enzyme acting in early steps of sterol isoprenoids biosynthesis, such as cholesterol in humans or ergosterol in trypanosomatids. MVK is conserved from bacteria to mammals, and localizes to glycosomes in trypanosomatids. During the course of T. cruzi MVK characterization, we found that, in addition to glycosomes, this enzyme may be secreted and modulate cell invasion. To evaluate the role of TcMVK in parasite-host cell interactions, TcMVK recombinant protein was produced and anti-TcMVK antibodies were raised in mice. TcMVK protein was detected in the supernatant of cultures of metacyclic trypomastigotes (MTs) and extracellular amastigotes (EAs) by Western blot analysis, confirming its secretion into extracellular medium. Recombinant TcMVK bound in a non-saturable dose-dependent manner to HeLa cells and positively modulated internalization of T. cruzi EAs but inhibited invasion by MTs. In HeLa cells, TcMVK induced phosphorylation of MAPK pathway components and proteins related to actin cytoskeleton modifications. We hypothesized that TcMVK is a bifunctional enzyme that in addition to playing a classical role in isoprenoid synthesis in glycosomes, it is secreted and may modulate host cell signaling required for T. cruzi invasion.
Collapse
Affiliation(s)
- Éden Ramalho Ferreira
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | | | - Alexis Bonfim-Melo
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Cristian Cortez
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Claudio Vieira da Silva
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | | | | | - Mário Costa Cruz
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Fabio Mitsuo Lima
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Esteban Mauricio Cordero
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Nobuko Yoshida
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - José Franco da Silveira
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Renato Arruda Mortara
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Diana Bahia
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil.,Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, MG, Brazil
| |
Collapse
|
27
|
Coelho C, Casadevall A. Cryptococcal therapies and drug targets: the old, the new and the promising. Cell Microbiol 2016; 18:792-9. [PMID: 26990050 DOI: 10.1111/cmi.12590] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 02/28/2016] [Accepted: 03/08/2016] [Indexed: 01/09/2023]
Abstract
Half a century after the introduction of Amphotericin B the management of cryptococcosis remains unsatisfactory. The disease, caused primarily by the two fungal species Cryptococcus neoformans and Cryptococcus gattii, remains responsible for considerable morbidity and mortality despite standard medical care. Current therapeutic options are limited to Amphotericin B, azoles and 5-flucytosine. However, this organism has numerous well-characterized virulence mechanisms that are amenable to pharmacological interference and are thus potential therapeutic targets. Here, we discuss existing approved antifungal drugs, resistance mechanisms to these drugs and non-standard antifungal drugs that have potential in treatment of cryptococcosis, including immunomodulatory strategies that synergize with antifungal drugs, such as cytokine administration or monoclonal antibodies. Finally, we summarize attempts to target well-described virulence factors of Cryptococcus, the capsule or fungal melanin. This review emphasizes the pressing need for new therapeutic alternatives for cryptococcosis.
Collapse
Affiliation(s)
- Carolina Coelho
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
28
|
Li X, Liu G, Ma J, Zhou L, Zhang Q, Gao L. Lack of IL-6 increases blood-brain barrier permeability in fungal meningitis. J Biosci 2015; 40:7-12. [PMID: 25740137 DOI: 10.1007/s12038-014-9496-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The pathogenesis of increased blood-brain barrier permeability during Cryptococcus meningitis is still largely unknown. Interleukin (IL-6) is a multifunctional cytokine, and numerous studies have shown that IL-6 influences the integrity of the blood-brain barrier. In this study we investigated the role of IL-6 in Cryptococcus meningitis. First, wild-type or IL-6(-/-) mice were injected with Cryptococcus neoformans (C. neoformans) and the survival time in both groups was recorded. Second, the number of fungi was measured in the brains of IL-6(-/-) wild-type mice. Finally, the blood-brain barrier permeability index was detected in infected IL-6(-/-) mice treated with recombinant human IL-6. The blood-brain barrier permeability index was measured in infected wild-type mice treated with anti-IL-6 antibodies as well. The survival of IL-6(-/-) mice injected with C. neoformans was significantly lower than that of identically challenged wild-type mice. The infected IL-6(-/-) mice had significantly larger brain fungal burdens than wild-type mice. Furthermore, increased blood-brain barrier index was found in infected IL-6(-/-) mice when compared with that in infected control mice. Similar results were obtained when mice challenged with C. neoformans were treated systemically with neutralizing anti-IL-6 antibodies, resulting in an elevation of vascular permeability. Our data revealed that IL-6 reduced the blood-brain barrier permeability during Cryptococcus meningitis, and it might provide an explanation for the significantly lower survival of infected IL-6(-/-) mice.
Collapse
Affiliation(s)
- Xiang Li
- Department of Pharmacy, First Affiliated Hospital of Chinese PLA General Hospital, Beijing 100048, China,
| | | | | | | | | | | |
Collapse
|
29
|
Atre AN, Mehta A, Chandorkar PR, Patole MS, Diwanay SS, Shah SR, Modak MS. Uptake of clinical yeast isolates by human epithelial cell line. J Mycol Med 2015; 26:187-92. [PMID: 26597144 DOI: 10.1016/j.mycmed.2015.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 10/01/2015] [Accepted: 10/02/2015] [Indexed: 10/22/2022]
Abstract
OBJECTIVE The occurrence of yeast infections in humans has increased, with the species belonging to genus Candida still being the most common cause of infection. Nevertheless, infections caused by less common yeasts have been widely reported in recent years. The main objective of this study was to assess the potential of these less common saprophytic yeasts to invade the host cell, which is essential for causing systemic infections. MATERIAL AND METHODS Various yeast isolates were identified by DNA sequence information of PCR amplified ITS region. The purported saprophytic yeasts were characterized for internalization by mammalian cells in vitro, by staining the F-actin. CONCLUSION The identification of different yeast isolates from various patients revealed that 70% of the isolates belonged to the genus Candida, while remaining 30% of the isolates were yeasts not belonging to genus Candida. These non-Candida clinical isolates, either in yeast or hyphal forms, were efficiently internalized by human epithelial cells. The internalization was marked by a process of actin polymerization surrounding the invading yeast. Such uptake by epithelial cells signifies traversal of cell barrier by yeast cells during infection in vivo.
Collapse
Affiliation(s)
- A N Atre
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Ganesh Khind, Pune 411007, India
| | - A Mehta
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Ganesh Khind, Pune 411007, India
| | - P R Chandorkar
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Ganesh Khind, Pune 411007, India
| | - M S Patole
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Ganesh Khind, Pune 411007, India.
| | - S S Diwanay
- M.E.S. Abasaheb Garware College, Karve Road, Pune 411004, India
| | - S R Shah
- Bharati Vidyapeeth's Medical College, Pune 411043, India
| | - M S Modak
- Bharati Vidyapeeth's Medical College, Pune 411043, India
| |
Collapse
|
30
|
Tseng HK, Huang TY, Wu AYJ, Chen HH, Liu CP, Jong A. How Cryptococcus interacts with the blood-brain barrier. Future Microbiol 2015; 10:1669-82. [PMID: 26437710 DOI: 10.2217/fmb.15.83] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Cryptococcus demonstrates predilection for invasion of the brain, but the mechanism by which Cryptococcus crosses the blood-brain barrier (BBB) to cause brain invasion is largely unknown. In order for Cryptococcus to cross the BBB, there must be a way to either cross human brain microvascular endothelial cells, which are the main constitute of the BBB, or go in between tight junctions. Recent evidence of human brain microvascular endothelial cell responses to transcellular brain invasions includes membrane rearrangements, intracellular signaling pathways and cytoskeletal activations. Several Cryptococcal genes related to the traversal of BBB have been identified, including CPS1, ITR1a, ITR3c, PLB1, MPR1, FNX1 and RUB1. In addition, Cryptococcus neoformans-derived microvesicles may contribute to cryptococcal brain invasion. Paracellularly, Cryptococcus may traverse across BBB using either routes utilizing plasmin, ammonia or macrophages in a Trojan horse mechanism.
Collapse
Affiliation(s)
- Hsiang-Kuang Tseng
- Department of Medicine, MacKay Medical College, No. 46, Sec. 3, Zhongzheng Road, Sanzhi Distric, New Taipei City 25245, Taiwan.,Division of Infectious Diseases, Department of Internal Medicine, MacKay Memorial Hospital, Taipei Branch, No. 92, Sec. 2, Zhongshan North Road, Taipei City 10449, Taiwan.,Microbiology Section, Department of Medical Research, MacKay Memorial Hospital, Tamshui Branch, No. 45, Minsheng Road, Tamshui District, New Taipei City 25160, Taiwan
| | - Tseng-Yu Huang
- Division of Infectious Diseases, Department of Internal Medicine, MacKay Memorial Hospital, Taipei Branch, No. 92, Sec. 2, Zhongshan North Road, Taipei City 10449, Taiwan
| | - Alice Ying-Jung Wu
- Division of Infectious Diseases, Department of Internal Medicine, MacKay Memorial Hospital, Taipei Branch, No. 92, Sec. 2, Zhongshan North Road, Taipei City 10449, Taiwan
| | - Hsin-Hong Chen
- Microbiology Section, Department of Medical Research, MacKay Memorial Hospital, Tamshui Branch, No. 45, Minsheng Road, Tamshui District, New Taipei City 25160, Taiwan
| | - Chang-Pan Liu
- Department of Medicine, MacKay Medical College, No. 46, Sec. 3, Zhongzheng Road, Sanzhi Distric, New Taipei City 25245, Taiwan.,Division of Infectious Diseases, Department of Internal Medicine, MacKay Memorial Hospital, Taipei Branch, No. 92, Sec. 2, Zhongshan North Road, Taipei City 10449, Taiwan.,Microbiology Section, Department of Medical Research, MacKay Memorial Hospital, Tamshui Branch, No. 45, Minsheng Road, Tamshui District, New Taipei City 25160, Taiwan
| | - Ambrose Jong
- Hematology-Oncology/BMT, Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| |
Collapse
|
31
|
Williams DW, Tesfa L, Berman JW. Novel flow cytometric analysis of the blood-brain barrier. Cytometry A 2015; 87:897-907. [PMID: 25929817 PMCID: PMC4859441 DOI: 10.1002/cyto.a.22683] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Revised: 03/20/2015] [Accepted: 04/11/2015] [Indexed: 12/13/2022]
Abstract
The blood-brain barrier (BBB) is primarily comprised of brain microvascular endothelial cells (BMVEC) and astrocytes and serves as a physical and chemical barrier that separates the periphery from the brain. We describe a flow cytometric method using our in vitro model of the human BBB to characterize BMVEC surface junctional proteins critical for maintenance of barrier function, cell viability, and leukocyte adhesion. For this methodology, BMVEC are cocultured with astrocytes in a transwell tissue culture insert to establish the barrier, after which time the BBB are treated with specific agents, and the BMVEC collected for flow cytometric analyses. We use a standard and optimized method to recover the BMVEC from the coculture model that maintains junctional protein expression and cell viability. A novel leukocyte adhesion assay enables a quantitative analysis of peripheral blood mononuclear cell (PBMC) interactions with the BMVEC and can be used to assess the adhesion of many cell types to the BBB. Furthermore, this method enables the concomitant analysis of a large number of adhesion molecules and tight junction proteins on both the BMVEC and adherent PBMC under homeostatic and pathologic conditions. Flow cytometry is an extremely powerful tool, and this technique can also be applied to assess variables not performed in this study, including cell cycle progression, and calcium flux.
Collapse
Affiliation(s)
- Dionna W. Williams
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, 10461
| | - Lydia Tesfa
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, 10461
| | - Joan W. Berman
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, 10461
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, 10461
| |
Collapse
|
32
|
Ueno N, Lodoen MB. From the blood to the brain: avenues of eukaryotic pathogen dissemination to the central nervous system. Curr Opin Microbiol 2015; 26:53-9. [PMID: 26048316 PMCID: PMC10538213 DOI: 10.1016/j.mib.2015.05.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 05/07/2015] [Accepted: 05/08/2015] [Indexed: 12/25/2022]
Abstract
Infection of the central nervous system (CNS) is a significant cause of morbidity and mortality, and treatments available to combat the highly debilitating symptoms of CNS infection are limited. The mechanisms by which pathogens in the circulation overcome host immunity and breach the blood-brain barrier are active areas of investigation. In this review, we discuss recent work that has significantly advanced our understanding of the avenues of pathogen dissemination to the CNS for four eukaryotic pathogens of global health importance: Toxoplasma gondii, Plasmodium falciparum, Trypanosoma brucei, and Cryptococcus neoformans. These studies highlight the remarkable diversity of pathogen strategies for trafficking to the brain and will ultimately contribute to an improved ability to combat life-threatening CNS disease.
Collapse
Affiliation(s)
- Norikiyo Ueno
- Department of Molecular Biology and Biochemistry and the Institute for Immunology, University of California, Irvine, CA, USA
| | - Melissa B Lodoen
- Department of Molecular Biology and Biochemistry and the Institute for Immunology, University of California, Irvine, CA, USA.
| |
Collapse
|
33
|
Yaeger R, Cowell E, Chou JF, Gewirtz AN, Borsu L, Vakiani E, Solit DB, Rosen N, Capanu M, Ladanyi M, Kemeny N. RAS mutations affect pattern of metastatic spread and increase propensity for brain metastasis in colorectal cancer. Cancer 2014; 121:1195-203. [PMID: 25491172 DOI: 10.1002/cncr.29196] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 11/03/2014] [Accepted: 11/17/2014] [Indexed: 12/25/2022]
Abstract
BACKGROUND RAS and PIK3CA mutations in metastatic colorectal cancer (mCRC) have been associated with worse survival. We sought to evaluate the impact of RAS and PIK3CA mutations on cumulative incidence of metastasis to potentially curable sites of liver and lung and other sites such as bone and brain. METHODS We performed a computerized search of the electronic medical record of our institution for mCRC cases genotyped for RAS or PIK3CA mutations from 2008 to 2012. Cases were reviewed for patient characteristics, survival, and site-specific metastasis. RESULTS Among the 918 patients identified, 477 cases were RAS wild type, and 441 cases had a RAS mutation (394 at KRAS exon 2, 29 at KRAS exon 3 or 4, and 18 in NRAS). RAS mutation was significantly associated with shorter median overall survival (OS) and on multivariate analysis independently predicted worse OS (HR, 1.6; P < .01). RAS mutant mCRC exhibited a significantly higher cumulative incidence of lung, bone, and brain metastasis and on multivariate analysis was an independent predictor of involvement of these sites (HR, 1.5, 1.6, and 3.7, respectively). PIK3CA mutations occurred in 10% of the 786 cases genotyped, did not predict for worse survival, and did not exhibit a site-specific pattern of metastatic spread. CONCLUSIONS The metastatic potential of CRC varies with the presence of RAS mutation. RAS mutation is associated with worse OS and increased incidence of lung, bone, and brain metastasis. An understanding of this site-specific pattern of spread may help to inform physicians' assessment of symptoms in patients with mCRC.
Collapse
Affiliation(s)
- Rona Yaeger
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Coelho C, Bocca AL, Casadevall A. The tools for virulence of Cryptococcus neoformans. ADVANCES IN APPLIED MICROBIOLOGY 2014; 87:1-41. [PMID: 24581388 DOI: 10.1016/b978-0-12-800261-2.00001-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cryptococcus neoformans is a fungal pathogen that causes almost half a million deaths each year. It is believed that most humans are infected with C. neoformans, possibly in a form that survives through latency in the lung and can reactivate to cause disease if the host becomes immunosuppressed. C. neoformans has a remarkably sophisticated intracellular survival capacities yet it is a free-living fungus with no requirement for mammalian virulence whatsoever. In this review, we discuss the tools that C. neoformans possesses to achieve survival, latency and virulence within its host. Some of these tools are mechanisms to withstand starvation and others aim to protect against microbicidal molecules produced by the immune system. Furthermore, we discuss how these tools were acquired through evolutionary pressures and perhaps accidental stochastic events, all of which combined to produce an organism with an unusual and unique intracellular pathogenic strategy.
Collapse
Affiliation(s)
- Carolina Coelho
- Department of Microbiology and Immunology, Albert Einstein College of Medicine of Yeshiva University, New York, USA; Centre for Neuroscience and Cell Biology of Coimbra, Institute of Microbiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Anamelia Lorenzetti Bocca
- Department of Cellular Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Arturo Casadevall
- Department of Microbiology and Immunology, Albert Einstein College of Medicine of Yeshiva University, New York, USA.
| |
Collapse
|
35
|
Invasion of the central nervous system by Cryptococcus neoformans requires a secreted fungal metalloprotease. mBio 2014; 5:e01101-14. [PMID: 24895304 PMCID: PMC4049100 DOI: 10.1128/mbio.01101-14] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
UNLABELLED Cryptococcus spp. cause life-threatening fungal infection of the central nervous system (CNS), predominantly in patients with a compromised immune system. Why Cryptococcus neoformans has this remarkable tropism for the CNS is not clear. Recent research on cerebral pathogenesis of C. neoformans revealed a predominantly transcellular migration of cryptococci across the brain endothelium; however, the identities of key fungal virulence factors that function specifically to invade the CNS remain unresolved. Here we found that a novel, secreted metalloprotease (Mpr1) that we identified in the extracellular proteome of C. neoformans (CnMpr1) is required for establishing fungal disease in the CNS. Mpr1 belongs to a poorly characterized M36 class of fungalysins that are expressed in only some fungal species. A strain of C. neoformans lacking the gene encoding Mpr1 (mpr1Δ) failed to breach the endothelium in an in vitro model of the human blood-brain barrier (BBB). A mammalian host infected with the mpr1Δ null strain demonstrated significant improvement in survival due to a reduced brain fungal burden and lacked the brain pathology commonly associated with cryptococcal disease. The in vivo studies further indicate that Mpr1 is not required for fungal dissemination and Mpr1 likely targets the brain endothelium specifically. Remarkably, the sole expression of CnMPR1 in Saccharomyces cerevisiae resulted in a robust migration of yeast cells across the brain endothelium, demonstrating Mpr1's specific activity in breaching the BBB and suggesting that Mpr1 may function independently of the hyaluronic acid-CD44 pathway. This distinct role for Mpr1 may develop into innovative treatment options and facilitate a brain-specific drug delivery platform. IMPORTANCE Cryptococcus neoformans is a medically relevant fungal pathogen causing significant morbidity and mortality, particularly in immunocompromised individuals. An intriguing feature is its strong neurotropism, and consequently the hallmark of cryptococcal disease is a brain infection, cryptococcal meningoencephalitis. For C. neoformans to penetrate the central nervous system (CNS), it first breaches the blood-brain barrier via a transcellular pathway; however, the identities of fungal factors required for this transmigration remain largely unknown. In an effort to identify extracellular fungal proteins that could mediate interactions with the brain endothelium, we undertook a proteomic analysis of the extracellular proteome and identified a secreted metalloprotease (Mpr1) belonging to the M36 class of fungalysins. Here we found that Mpr1 promotes migration of C. neoformans across the brain endothelium and into the CNS by facilitating attachment of cryptococci to the endothelium surface, thus underscoring the critical role of M36 proteases in fungal pathogenesis.
Collapse
|
36
|
The involvement of FAK and Src in the invasion of cardiomyocytes by Trypanosoma cruzi. Exp Parasitol 2014; 139:49-57. [PMID: 24582948 DOI: 10.1016/j.exppara.2014.02.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 12/10/2013] [Accepted: 02/05/2014] [Indexed: 11/20/2022]
Abstract
The activation of signaling pathways involving protein tyrosine kinases (PTKs) has been demonstrated during Trypanosoma cruzi invasion. Herein, we describe the participation of FAK/Src in the invasion of cardiomyocytes by T. cruzi. The treatment of cardiomyocytes with genistein, a PTK inhibitor, significantly reduced T. cruzi invasion. Also, PP1, a potent Src-family protein inhibitor, and PF573228, a specific FAK inhibitor, also inhibited T. cruzi entry; maximal inhibition was achieved at concentrations of 25μM PP1 (53% inhibition) and 40μM PF573228 (50% inhibition). The suppression of FAK expression in siRNA-treated cells and tetracycline-uninduced Tet-FAK(WT)-46 cells significantly reduced T. cruzi invasion. The entry of T. cruzi is accompanied by changes in FAK and c-Src expression and phosphorylation. An enhancement of FAK activation occurs during the initial stages of T. cruzi-cardiomyocyte interaction (30 and 60min), with a concomitant increase in the level of c-Src expression and phosphorylation, suggesting that FAK/Src act as an integrated signaling pathway that coordinates parasite entry. These data provide novel insights into the signaling pathways that are involved in cardiomyocyte invasion by T. cruzi. A better understanding of the signal transduction networks involved in T. cruzi invasion may contribute to the development of more effective therapies for the treatment of Chagas' disease.
Collapse
|
37
|
Liu TB, Kim JC, Wang Y, Toffaletti DL, Eugenin E, Perfect JR, Kim KJ, Xue C. Brain inositol is a novel stimulator for promoting Cryptococcus penetration of the blood-brain barrier. PLoS Pathog 2013; 9:e1003247. [PMID: 23592982 PMCID: PMC3617100 DOI: 10.1371/journal.ppat.1003247] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 02/01/2013] [Indexed: 11/17/2022] Open
Abstract
Cryptococcus neoformans is the most common cause of fungal meningitis, with high mortality and morbidity. The reason for the frequent occurrence of Cryptococcus infection in the central nervous system (CNS) is poorly understood. The facts that human and animal brains contain abundant inositol and that Cryptococcus has a sophisticated system for the acquisition of inositol from the environment suggests that host inositol utilization may contribute to the development of cryptococcal meningitis. In this study, we found that inositol plays an important role in Cryptococcus traversal across the blood-brain barrier (BBB) both in an in vitro human BBB model and in in vivo animal models. The capacity of inositol to stimulate BBB crossing was dependent upon fungal inositol transporters, indicated by a 70% reduction in transmigration efficiency in mutant strains lacking two major inositol transporters, Itr1a and Itr3c. Upregulation of genes involved in the inositol catabolic pathway was evident in a microarray analysis following inositol treatment. In addition, inositol increased the production of hyaluronic acid in Cryptococcus cells, which is a ligand known to binding host CD44 receptor for their invasion. These studies suggest an inositol-dependent Cryptococcus traversal of the BBB, and support our hypothesis that utilization of host-derived inositol by Cryptococcus contributes to CNS infection. Cryptococcus neoformans is an AIDS-associated human fungal pathogen that annually causes over 1 million cases of meningitis world-wide, and more than 600,000 attributable deaths. Cryptococcus often causes lung and brain infection and is the leading cause of fungal meningitis in immunosuppressed patients. Why Cryptococcus frequently infects the central nervous system to cause fatal meningitis is an unanswered critical question. Our previous studies revealed a sophisticated inositol acquisition system in Cryptococcus that plays a central role in utilizing environmental inositol to complete its sexual cycle. Here we further demonstrate that inositol acquisition is also important for fungal infection in the brain, where abundant inositol is available. We found that inositol promotes the traversal of Cryptococcus across the blood-brain barrier (BBB), and such stimulation is fungal inositol transporter dependent. We also identified the effects of host inositol on fungal cellular functions that contribute to the stimulation of fungal penetration of the BBB. We propose that inositol utilization is a novel virulence factor for CNS cryptococcosis. Our work lays an important foundation for understanding how fungi respond to available host inositol and indicates the impact of host inositol acquisition on the development of cryptococcal meningitis.
Collapse
Affiliation(s)
- Tong-Bao Liu
- Public Health Research Institute Center, University of Medicine and Dentistry of New Jersey, Newark, New Jersey, United States of America
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Lu Z, Bast RC. The tumor suppressor gene ARHI (DIRAS3) inhibits ovarian cancer cell migration through multiple mechanisms. Cell Adh Migr 2013; 7:232-6. [PMID: 23357870 DOI: 10.4161/cam.23648] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
ARHI is an imprinted tumor suppressor gene that is downregulated in > 60% of ovarian cancers, associated with decreased progression-free survival. ARHI encodes a 26 kDa GTPase with homology to Ras. Re-expression of ARHI inhibits ovarian cancer growth, initiates autophagy and induces tumor dormancy. Recent studies have demonstrated that ARHI also plays a particularly important role in ovarian cancer cell migration. Re-expression of ARHI decreases motility of IL-6- and EGF-stimulated SKOv3 and Hey ovarian cancer cells, inhibiting both chemotaxis and haptotaxis. ARHI inhibits cell migration by binding and sequestering STAT3 in the cytoplasm, and preventing STAT3 translocation to the nucleus and localization in focal adhesion complexes. Re-expression of ARHI inhibits FAK (Y397) phosphorylation, disrupts focal adhesions and blocks FAK-mediated RhoA signaling, resulting in decreased levels of GTP-RhoA. Re-expression of ARHI disrupts formation of actin stress fibers in a FAK- and RhoA-dependent manner. Recent studies indicate that re-expression of ARHI inhibits expression of β-1 integrin which may also contribute to inhibition of migration, adhesion and invasion.
Collapse
Affiliation(s)
- Zhen Lu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | |
Collapse
|