1
|
Stone M, Choi CS, Dey N, Swain G, Stevens T, Sayner SL. Pseudomonas aeruginosa ExoY infection of pulmonary microvascular endothelial cells releases cyclic nucleotides into the extracellular compartment. Am J Physiol Lung Cell Mol Physiol 2024; 327:L756-L768. [PMID: 39316682 PMCID: PMC11560077 DOI: 10.1152/ajplung.00038.2024] [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: 02/05/2024] [Revised: 08/19/2024] [Accepted: 09/05/2024] [Indexed: 09/26/2024] Open
Abstract
Type three secretion system (TTSS)-competent Pseudomonas aeruginosa expressing soluble promiscuous cyclase, exoenzyme Y (ExoY), generates cyclic nucleotides in pulmonary microvascular endothelial cells (PMVECs). Within cells, cyclic nucleotide signals are highly compartmentalized, but these second messengers are also released into the extracellular space. Although agonist stimulation of endogenous adenylyl cyclase (AC) or the presence of ExoY increases cyclic nucleotides, the proportion of the signal that is in the intracellular versus extracellular compartments is unresolved. Furthermore, it is unclear whether P. aeruginosa primary infection or treatment with sterile media supernatants derived from a primary infection alters beta-adrenergic agonist-induced elevations in cAMP in PMVECs. Herein, we determine that PMVECs release cAMP into the extracellular space constitutively, following beta-adrenergic stimulation of endogenous AC, and following infection with P. aeruginosa expressing ExoY. Surprisingly, in PMVECs, only a small proportion of cGMP is detected within the cell at baseline or following P. aeruginosa ExoY infection with a larger proportion of total cGMP being detected extracellularly. Thus, the ability of lung endothelium to generate cyclic nucleotides may be underestimated by examining intracellular cyclic nucleotides alone, since a large portion is delivered into the extracellular compartment. In addition, P. aeruginosa infection or treatment with sterile media supernatants from a primary infection suppresses the beta-adrenergic cAMP response, which is further attenuated by the expression of functional ExoY. These findings reveal an overabundance of extracellular cyclic nucleotides following infection with ExoY expressing TTSS-competent P. aeruginosa.NEW & NOTEWORTHY P. aeruginosa exoenzyme Y (ExoY) infection increases cyclic nucleotides intracellularly, but an overabundance of cAMP and cGMP is also detected in the extracellular space and reveals a greater capacity of pulmonary endothelial cells to generate cAMP and cGMP. P. aeruginosa infection or treatment with sterile media supernatants derived from a primary infection suppresses the β-adrenergic-induced cAMP response in pulmonary endothelial cells, which is exacerbated by the expression of functional ExoY.
Collapse
Affiliation(s)
- Madeline Stone
- Department of Physiology and Cell Biology, University South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University South Alabama, Mobile, Alabama, United States
| | - Chung-Sik Choi
- Department of Physiology and Cell Biology, University South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University South Alabama, Mobile, Alabama, United States
| | - Nandita Dey
- Department of Physiology and Cell Biology, University South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University South Alabama, Mobile, Alabama, United States
| | - Grace Swain
- Department of Physiology and Cell Biology, University South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University South Alabama, Mobile, Alabama, United States
| | - Troy Stevens
- Department of Physiology and Cell Biology, University South Alabama, Mobile, Alabama, United States
- Department of Internal Medicine, University South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University South Alabama, Mobile, Alabama, United States
| | - Sarah L Sayner
- Department of Physiology and Cell Biology, University South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University South Alabama, Mobile, Alabama, United States
| |
Collapse
|
2
|
deWeever A, Paudel SS, Zhou C, Francis CM, Tambe DT, Frank DW, Balczon R, Stevens T. cUMP elicits interendothelial gap formation during Pseudomonas aeruginosa infection. Am J Physiol Lung Cell Mol Physiol 2024; 327:L395-L405. [PMID: 39076085 PMCID: PMC11444506 DOI: 10.1152/ajplung.00164.2023] [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: 05/22/2023] [Revised: 05/08/2024] [Accepted: 06/30/2024] [Indexed: 07/31/2024] Open
Abstract
Pseudomonas aeruginosa utilizes a type 3 secretion system to intoxicate host cells with the nucleotidyl cyclase ExoY. After activation by its host cell cofactor, filamentous actin, ExoY produces purine and pyrimidine cyclic nucleotides, including cAMP, cGMP, and cUMP. ExoY-generated cyclic nucleotides promote interendothelial gap formation, impair motility, and arrest cell growth. The disruptive activities of cAMP and cGMP during the P. aeruginosa infection are established; however, little is known about the function of cUMP. Here, we tested the hypothesis that cUMP contributes to endothelial cell barrier disruption during P. aeruginosa infection. Using a membrane permeable cUMP analog, cUMP-AM, we revealed that during infection with catalytically inactive ExoY, cUMP promotes interendothelial gap formation in cultured pulmonary microvascular endothelial cells (PMVECs) and contributes to increased filtration coefficient in the isolated perfused lung. These findings indicate that cUMP contributes to endothelial permeability during P. aeruginosa lung infection.NEW & NOTEWORTHY During pneumonia, bacteria utilize a virulence arsenal to communicate with host cells. The Pseudomonas aeruginosa T3SS directly introduces virulence molecules into the host cell cytoplasm. These molecules are enzymes that trigger interkingdom communication. One of the exoenzymes is a nucleotidyl cyclase that produces noncanonical cyclic nucleotides like cUMP. Little is known about how cUMP acts in the cell. Here we found that cUMP instigates pulmonary edema during Pseudomonas aeruginosa infection of the lung.
Collapse
Grants
- R01 HL167997 NHLBI NIH HHS
- HL136689 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- P01 HL066299 NHLBI NIH HHS
- AI104922 HHS | NIH | NIAID | Division of Microbiology and Infectious Diseases (DMID)
- R01 HL140182 NHLBI NIH HHS
- HL167997 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 AI104922 NIAID NIH HHS
- HL148069 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL148069 NHLBI NIH HHS
- HL140182 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL66299 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
Collapse
Affiliation(s)
- Althea deWeever
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Sunita S Paudel
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Chun Zhou
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - C Michael Francis
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Dhananjay T Tambe
- Department of Mechanical, Aerospace and Biomedical Engineering, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Dara W Frank
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Center for Infectious Disease Research, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Ron Balczon
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Troy Stevens
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Department of Internal Medicine, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| |
Collapse
|
3
|
Balczon R, Lin MT, Voth S, Nelson AR, Schupp JC, Wagener BM, Pittet JF, Stevens T. Lung endothelium, tau, and amyloids in health and disease. Physiol Rev 2024; 104:533-587. [PMID: 37561137 PMCID: PMC11281824 DOI: 10.1152/physrev.00006.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 06/26/2023] [Accepted: 08/04/2023] [Indexed: 08/11/2023] Open
Abstract
Lung endothelia in the arteries, capillaries, and veins are heterogeneous in structure and function. Lung capillaries in particular represent a unique vascular niche, with a thin yet highly restrictive alveolar-capillary barrier that optimizes gas exchange. Capillary endothelium surveys the blood while simultaneously interpreting cues initiated within the alveolus and communicated via immediately adjacent type I and type II epithelial cells, fibroblasts, and pericytes. This cell-cell communication is necessary to coordinate the immune response to lower respiratory tract infection. Recent discoveries identify an important role for the microtubule-associated protein tau that is expressed in lung capillary endothelia in the host-pathogen interaction. This endothelial tau stabilizes microtubules necessary for barrier integrity, yet infection drives production of cytotoxic tau variants that are released into the airways and circulation, where they contribute to end-organ dysfunction. Similarly, beta-amyloid is produced during infection. Beta-amyloid has antimicrobial activity, but during infection it can acquire cytotoxic activity that is deleterious to the host. The production and function of these cytotoxic tau and amyloid variants are the subject of this review. Lung-derived cytotoxic tau and amyloid variants are a recently discovered mechanism of end-organ dysfunction, including neurocognitive dysfunction, during and in the aftermath of infection.
Collapse
Affiliation(s)
- Ron Balczon
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Mike T Lin
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Sarah Voth
- Department of Cell Biology and Physiology, Edward Via College of Osteopathic Medicine, Monroe, Louisiana, United States
| | - Amy R Nelson
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Jonas C Schupp
- Pulmonary and Critical Care Medicine, Department of Internal Medicine, Yale University, New Haven, Connecticut, United States
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
- German Center for Lung Research (DZL), Hannover, Germany
| | - Brant M Wagener
- Department of Anesthesiology and Perioperative Medicine, University of Alabama-Birmingham, Birmingham, Alabama, United States
| | - Jean-Francois Pittet
- Department of Anesthesiology and Perioperative Medicine, University of Alabama-Birmingham, Birmingham, Alabama, United States
| | - Troy Stevens
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Department of Internal Medicine, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| |
Collapse
|
4
|
Walker A, Czyz DM. Oh my gut! Is the microbial origin of neurodegenerative diseases real? Infect Immun 2023; 91:e0043722. [PMID: 37750713 PMCID: PMC10580905 DOI: 10.1128/iai.00437-22] [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: 09/27/2023] Open
Abstract
There is no cure or effective treatment for neurodegenerative protein conformational diseases (PCDs), such as Alzheimer's or Parkinson's diseases, mainly because the etiology of these diseases remains elusive. Recent data suggest that unique changes in the gut microbial composition are associated with these ailments; however, our current understanding of the bacterial role in the pathogenesis of PCDs is hindered by the complexity of the microbial communities associated with specific microbiomes, such as the gut, oral, or vaginal microbiota. The composition of these specific microbiomes is regarded as a unique fingerprint affected by factors such as infections, diet, lifestyle, and antibiotics. All of these factors also affect the severity of neurodegenerative diseases. The majority of studies that reveal microbial contribution are correlational, and various models, including worm, fly, and mouse, are being utilized to decipher the role of individual microbes that may affect disease onset and progression. Recent evidence from across model organisms and humans shows a positive correlation between the presence of gram-negative enteropathogenic bacteria and the pathogenesis of PCDs. While these correlational studies do not provide a mechanistic explanation, they do reveal contributing bacterial species and provide an important basis for further investigation. One of the lurking concerns related to the microbial contribution to PCDs is the increasing prevalence of antibiotic resistance and poor antibiotic stewardship, which ultimately select for proteotoxic bacteria, especially the gram-negative species that are known for intrinsic resistance. In this review, we summarize what is known about individual microbial contribution to PCDs and the potential impact of increasing antimicrobial resistance.
Collapse
Affiliation(s)
- Alyssa Walker
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, USA
| | - Daniel M. Czyz
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
5
|
Gwin MS, Alexeyev MF, Geurts AM, Lee JY, Zhou C, Yang XM, Cohen MV, Downey JM, Barrington RA, Spadafora D, Audia JP, Frank DW, Voth S, Pastukh VV, Bell J, Ayers L, Tambe DT, Nelson AR, Balczon R, Lin MT, Stevens T. Gamma secretase activating protein promotes end-organ dysfunction after bacterial pneumonia. Am J Physiol Lung Cell Mol Physiol 2023; 325:L174-L189. [PMID: 37366533 PMCID: PMC10396227 DOI: 10.1152/ajplung.00018.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 06/20/2023] [Accepted: 06/20/2023] [Indexed: 06/28/2023] Open
Abstract
Pneumonia elicits the production of cytotoxic beta amyloid (Aβ) that contributes to end-organ dysfunction, yet the mechanism(s) linking infection to activation of the amyloidogenic pathway that produces cytotoxic Aβ is unknown. Here, we tested the hypothesis that gamma-secretase activating protein (GSAP), which contributes to the amyloidogenic pathway in the brain, promotes end-organ dysfunction following bacterial pneumonia. First-in-kind Gsap knockout rats were generated. Wild-type and knockout rats possessed similar body weights, organ weights, circulating blood cell counts, arterial blood gases, and cardiac indices at baseline. Intratracheal Pseudomonas aeruginosa infection caused acute lung injury and a hyperdynamic circulatory state. Whereas infection led to arterial hypoxemia in wild-type rats, the alveolar-capillary barrier integrity was preserved in Gsap knockout rats. Infection potentiated myocardial infarction following ischemia-reperfusion injury, and this potentiation was abolished in knockout rats. In the hippocampus, GSAP contributed to both pre- and postsynaptic neurotransmission, increasing the presynaptic action potential recruitment, decreasing neurotransmitter release probability, decreasing the postsynaptic response, and preventing postsynaptic hyperexcitability, resulting in greater early long-term potentiation but reduced late long-term potentiation. Infection abolished early and late long-term potentiation in wild-type rats, whereas the late long-term potentiation was partially preserved in Gsap knockout rats. Furthermore, hippocampi from knockout rats, and both the wild-type and knockout rats following infection, exhibited a GSAP-dependent increase in neurotransmitter release probability and postsynaptic hyperexcitability. These results elucidate an unappreciated role for GSAP in innate immunity and highlight the contribution of GSAP to end-organ dysfunction during infection.NEW & NOTEWORTHY Pneumonia is a common cause of end-organ dysfunction, both during and in the aftermath of infection. In particular, pneumonia is a common cause of lung injury, increased risk of myocardial infarction, and neurocognitive dysfunction, although the mechanisms responsible for such increased risk are unknown. Here, we reveal that gamma-secretase activating protein, which contributes to the amyloidogenic pathway, is important for end-organ dysfunction following infection.
Collapse
Affiliation(s)
- Meredith S Gwin
- Department of Physiology and Cell Biology, Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Mikhail F Alexeyev
- Department of Physiology and Cell Biology, Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Aron M Geurts
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Ji Young Lee
- Department of Physiology and Cell Biology, Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Chun Zhou
- Department of Physiology and Cell Biology, Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Xi-Ming Yang
- Department of Physiology and Cell Biology, Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Michael V Cohen
- Department of Physiology and Cell Biology, Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - James M Downey
- Department of Physiology and Cell Biology, Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Robert A Barrington
- Department of Microbiology and Immunology, Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Domenico Spadafora
- Department of Microbiology and Immunology, Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Jonathon P Audia
- Department of Microbiology and Immunology, Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Dara W Frank
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Sarah Voth
- Department of Cell Biology and Physiology, Edward Via College of Osteopathic Medicine, Monroe, Louisiana, United States
| | - Viktoriya V Pastukh
- Department of Physiology and Cell Biology, Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Jessica Bell
- Department of Physiology and Cell Biology, Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Linn Ayers
- Department of Physiology and Cell Biology, Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Dhananjay T Tambe
- Department of Mechanical, Aerospace, and Biomedical Engineering, Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Amy R Nelson
- Department of Physiology and Cell Biology, Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Ron Balczon
- Department of Biochemistry and Molecular Biology, Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Mike T Lin
- Department of Physiology and Cell Biology, Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Troy Stevens
- Department of Physiology and Cell Biology, Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| |
Collapse
|
6
|
Balczon R, Choi CS, deWeever A, Zhou C, Gwin MS, Kolb C, Francis CM, Lin MT, Stevens T. Infection promotes Ser-214 phosphorylation important for generation of cytotoxic tau variants. FASEB J 2023; 37:e23042. [PMID: 37358817 DOI: 10.1096/fj.202300620rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/02/2023] [Accepted: 06/07/2023] [Indexed: 06/27/2023]
Abstract
Patients who recover from hospital-acquired pneumonia exhibit a high incidence of end-organ dysfunction following hospital discharge, including cognitive deficits. We have previously demonstrated that pneumonia induces the production and release of cytotoxic oligomeric tau from pulmonary endothelial cells, and these tau oligomers can enter the circulation and may be a cause of long-term morbidities. Endothelial-derived oligomeric tau is hyperphosphorylated during infection. The purpose of these studies was to determine whether Ser-214 phosphorylation of tau is a necessary stimulus for generation of cytotoxic tau variants. The results of these studies demonstrate that Ser-214 phosphorylation is critical for the cytotoxic properties of infection-induced oligomeric tau. In the lung, Ser-214 phosphorylated tau contributes to disruption of the alveolar-capillary barrier, resulting in increased permeability. However, in the brain, both the Ser-214 phosphorylated tau and the mutant Ser-214-Ala tau, which cannot be phosphorylated, disrupted hippocampal long-term potentiation suggesting that inhibition of long-term potentiation was relatively insensitive to the phosphorylation status of Ser-214. Nonetheless, phosphorylation of tau is essential to its cytotoxicity since global dephosphorylation of the infection-induced cytotoxic tau variants rescued long-term potentiation. Collectively, these data demonstrate that multiple forms of oligomeric tau are generated during infectious pneumonia, with different forms of oligomeric tau being responsible for dysfunction of distinct end-organs during pneumonia.
Collapse
Affiliation(s)
- Ron Balczon
- Department of Biochemistry and Molecular Biology, University of South Alabama College of Medicine, Mobile, Alabama, USA
- Center for Lung Biology, University of South Alabama College of Medicine, Mobile, Alabama, USA
| | - Chung-Sik Choi
- Center for Lung Biology, University of South Alabama College of Medicine, Mobile, Alabama, USA
- Department of Physiology and Cell Biology, University of South Alabama College of Medicine, Mobile, Alabama, USA
| | - Althea deWeever
- Center for Lung Biology, University of South Alabama College of Medicine, Mobile, Alabama, USA
- Department of Physiology and Cell Biology, University of South Alabama College of Medicine, Mobile, Alabama, USA
| | - Chun Zhou
- Center for Lung Biology, University of South Alabama College of Medicine, Mobile, Alabama, USA
- Department of Physiology and Cell Biology, University of South Alabama College of Medicine, Mobile, Alabama, USA
| | - Meredith S Gwin
- Center for Lung Biology, University of South Alabama College of Medicine, Mobile, Alabama, USA
- Department of Physiology and Cell Biology, University of South Alabama College of Medicine, Mobile, Alabama, USA
| | - Claire Kolb
- Department of Biochemistry and Molecular Biology, University of South Alabama College of Medicine, Mobile, Alabama, USA
- Center for Lung Biology, University of South Alabama College of Medicine, Mobile, Alabama, USA
| | - C Michael Francis
- Center for Lung Biology, University of South Alabama College of Medicine, Mobile, Alabama, USA
- Department of Physiology and Cell Biology, University of South Alabama College of Medicine, Mobile, Alabama, USA
| | - Mike T Lin
- Center for Lung Biology, University of South Alabama College of Medicine, Mobile, Alabama, USA
- Department of Physiology and Cell Biology, University of South Alabama College of Medicine, Mobile, Alabama, USA
| | - Troy Stevens
- Center for Lung Biology, University of South Alabama College of Medicine, Mobile, Alabama, USA
- Department of Physiology and Cell Biology, University of South Alabama College of Medicine, Mobile, Alabama, USA
| |
Collapse
|
7
|
Hussong SA, Banh AQ, Van Skike CE, Dorigatti AO, Hernandez SF, Hart MJ, Ferran B, Makhlouf H, Gaczynska M, Osmulski PA, McAllen SA, Dineley KT, Ungvari Z, Perez VI, Kayed R, Galvan V. Soluble pathogenic tau enters brain vascular endothelial cells and drives cellular senescence and brain microvascular dysfunction in a mouse model of tauopathy. Nat Commun 2023; 14:2367. [PMID: 37185259 PMCID: PMC10126555 DOI: 10.1038/s41467-023-37840-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 03/31/2023] [Indexed: 05/17/2023] Open
Abstract
Vascular mechanisms of Alzheimer's disease (AD) may constitute a therapeutically addressable biological pathway underlying dementia. We previously demonstrated that soluble pathogenic forms of tau (tau oligomers) accumulate in brain microvasculature of AD and other tauopathies, including prominently in microvascular endothelial cells. Here we show that soluble pathogenic tau accumulates in brain microvascular endothelial cells of P301S(PS19) mice modeling tauopathy and drives AD-like brain microvascular deficits. Microvascular impairments in P301S(PS19) mice were partially negated by selective removal of pathogenic soluble tau aggregates from brain. We found that similar to trans-neuronal transmission of pathogenic forms of tau, soluble tau aggregates are internalized by brain microvascular endothelial cells in a heparin-sensitive manner and induce microtubule destabilization, block endothelial nitric oxide synthase (eNOS) activation, and potently induce endothelial cell senescence that was recapitulated in vivo in microvasculature of P301S(PS19) mice. Our studies suggest that soluble pathogenic tau aggregates mediate AD-like brain microvascular deficits in a mouse model of tauopathy, which may arise from endothelial cell senescence and eNOS dysfunction triggered by internalization of soluble tau aggregates.
Collapse
Affiliation(s)
- Stacy A Hussong
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
- Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
- Oklahoma City Veterans Health Care System, 921 NE 13th Street, Oklahoma City, OK, 73104, USA
| | - Andy Q Banh
- South Texas Medical Scientist Training Program, University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, 4939 Charles Katz Drive, San Antonio, TX, 78229, USA
| | - Candice E Van Skike
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, 4939 Charles Katz Drive, San Antonio, TX, 78229, USA
| | - Angela O Dorigatti
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, 4939 Charles Katz Drive, San Antonio, TX, 78229, USA
| | - Stephen F Hernandez
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, 4939 Charles Katz Drive, San Antonio, TX, 78229, USA
| | - Matthew J Hart
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
- Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
- Center for Therapeutic Science, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
| | - Beatriz Ferran
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
- Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
| | - Haneen Makhlouf
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
- Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
| | - Maria Gaczynska
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, 4939 Charles Katz Drive, San Antonio, TX, 78229, USA
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | - Pawel A Osmulski
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, 4939 Charles Katz Drive, San Antonio, TX, 78229, USA
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | - Salome A McAllen
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA
- Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA
- Sealy Center for Vaccine Development, University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA
| | - Kelly T Dineley
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA
- Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA
- Sealy Center for Vaccine Development, University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA
| | - Zoltan Ungvari
- Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, 800 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine, Department of Public Health, Semmelweis University, H-1085 Budapest, Üllői út 26, Budapest, Hungary
| | | | - Rakez Kayed
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA
- Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA
- Sealy Center for Vaccine Development, University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA
| | - Veronica Galvan
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA.
- Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA.
- Oklahoma City Veterans Health Care System, 921 NE 13th Street, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
8
|
What the Gut Tells the Brain-Is There a Link between Microbiota and Huntington's Disease? Int J Mol Sci 2023; 24:ijms24054477. [PMID: 36901907 PMCID: PMC10003333 DOI: 10.3390/ijms24054477] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 02/14/2023] [Accepted: 02/20/2023] [Indexed: 03/12/2023] Open
Abstract
The human intestinal microbiota is a diverse and dynamic microenvironment that forms a complex, bi-directional relationship with the host. The microbiome takes part in the digestion of food and the generation of crucial nutrients such as short chain fatty acids (SCFA), but is also impacts the host's metabolism, immune system, and even brain functions. Due to its indispensable role, microbiota has been implicated in both the maintenance of health and the pathogenesis of many diseases. Dysbiosis in the gut microbiota has already been implicated in many neurodegenerative diseases such as Parkinson's disease (PD) and Alzheimer's disease (AD). However, not much is known about the microbiome composition and its interactions in Huntington's disease (HD). This dominantly heritable, incurable neurodegenerative disease is caused by the expansion of CAG trinucleotide repeats in the huntingtin gene (HTT). As a result, toxic RNA and mutant protein (mHTT), rich in polyglutamine (polyQ), accumulate particularly in the brain, leading to its impaired functions. Interestingly, recent studies indicated that mHTT is also widely expressed in the intestines and could possibly interact with the microbiota, affecting the progression of HD. Several studies have aimed so far to screen the microbiota composition in mouse models of HD and find out whether observed microbiome dysbiosis could affect the functions of the HD brain. This review summarizes ongoing research in the HD field and highlights the essential role of the intestine-brain axis in HD pathogenesis and progression. The review also puts a strong emphasis on indicating microbiome composition as a future target in the urgently needed therapy for this still incurable disease.
Collapse
|
9
|
Significance of Pulmonary Endothelial Injury and the Role of Cyclooxygenase-2 and Prostanoid Signaling. BIOENGINEERING (BASEL, SWITZERLAND) 2023; 10:bioengineering10010117. [PMID: 36671689 PMCID: PMC9855370 DOI: 10.3390/bioengineering10010117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023]
Abstract
The endothelium plays a key role in the dynamic balance of hemodynamic, humoral and inflammatory processes in the human body. Its central importance and the resulting therapeutic concepts are the subject of ongoing research efforts and form the basis for the treatment of numerous diseases. The pulmonary endothelium is an essential component for the gas exchange in humans. Pulmonary endothelial dysfunction has serious consequences for the oxygenation and the gas exchange in humans with the potential of consecutive multiple organ failure. Therefore, in this review, the dysfunction of the pulmonary endothel due to viral, bacterial, and fungal infections, ventilator-related injury, and aspiration is presented in a medical context. Selected aspects of the interaction of endothelial cells with primarily alveolar macrophages are reviewed in more detail. Elucidation of underlying causes and mechanisms of damage and repair may lead to new therapeutic approaches. Specific emphasis is placed on the processes leading to the induction of cyclooxygenase-2 and downstream prostanoid-based signaling pathways associated with this enzyme.
Collapse
|
10
|
Wood SJ, Goldufsky JW, Seu MY, Dorafshar AH, Shafikhani SH. Pseudomonas aeruginosa Cytotoxins: Mechanisms of Cytotoxicity and Impact on Inflammatory Responses. Cells 2023; 12:cells12010195. [PMID: 36611990 PMCID: PMC9818787 DOI: 10.3390/cells12010195] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/01/2022] [Accepted: 12/01/2022] [Indexed: 01/05/2023] Open
Abstract
Pseudomonas aeruginosa is one of the most virulent opportunistic Gram-negative bacterial pathogens in humans. It causes many acute and chronic infections with morbidity and mortality rates as high as 40%. P. aeruginosa owes its pathogenic versatility to a large arsenal of cell-associated and secreted virulence factors which enable this pathogen to colonize various niches within hosts and protect it from host innate immune defenses. Induction of cytotoxicity in target host cells is a major virulence strategy for P. aeruginosa during the course of infection. P. aeruginosa has invested heavily in this strategy, as manifested by a plethora of cytotoxins that can induce various forms of cell death in target host cells. In this review, we provide an in-depth review of P. aeruginosa cytotoxins based on their mechanisms of cytotoxicity and the possible consequences of their cytotoxicity on host immune responses.
Collapse
Affiliation(s)
- Stephen J. Wood
- Department of Medicine, Division of Hematology, Oncology and Cell Therapy, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Rush University Medical Center, Chicago, IL 60612, USA
| | - Josef W. Goldufsky
- Department of Medicine, Division of Hematology, Oncology and Cell Therapy, Rush University Medical Center, Chicago, IL 60612, USA
| | - Michelle Y. Seu
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Rush University Medical Center, Chicago, IL 60612, USA
| | - Amir H. Dorafshar
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Rush University Medical Center, Chicago, IL 60612, USA
| | - Sasha H. Shafikhani
- Department of Medicine, Division of Hematology, Oncology and Cell Therapy, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL 60612, USA
- Cancer Center, Rush University Medical Center, Chicago, IL 60612, USA
- Correspondence:
| |
Collapse
|
11
|
Davi M, Sadi M, Pitard I, Chenal A, Ladant D. A Robust and Sensitive Spectrophotometric Assay for the Enzymatic Activity of Bacterial Adenylate Cyclase Toxins. Toxins (Basel) 2022; 14:toxins14100691. [PMID: 36287960 PMCID: PMC9609896 DOI: 10.3390/toxins14100691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/06/2022] Open
Abstract
Various bacterial pathogens are producing toxins that target the cyclic Nucleotide Monophosphate (cNMPs) signaling pathways in order to facilitate host colonization. Among them, several are exhibiting potent nucleotidyl cyclase activities that are activated by eukaryotic factors, such as the adenylate cyclase (AC) toxin, CyaA, from Bordetella pertussis or the edema factor, EF, from Bacillus anthracis. The characterization of these toxins frequently requires accurate measurements of their enzymatic activity in vitro, in particular for deciphering their structure-to-function relationships by protein engineering and site-directed mutagenesis. Here we describe a simple and robust in vitro assay for AC activity based on the spectrophotometric detection of cyclic AMP (cAMP) after chromatographic separation on aluminum oxide. This assay can accurately detect down to fmol amounts of B. pertussis CyaA and can even be used in complex media, such as cell extracts. The relative advantages and disadvantages of this assay in comparison with other currently available methods are briefly discussed.
Collapse
Affiliation(s)
- Marilyne Davi
- Biochemistry of Macromolecular Interactions Unit, Department of Structural Biology and Chemistry, Institut Pasteur, Université Paris Cité, CNRS UMR 3528, 75015 Paris, France
| | - Mirko Sadi
- Biochemistry of Macromolecular Interactions Unit, Department of Structural Biology and Chemistry, Institut Pasteur, Université Paris Cité, CNRS UMR 3528, 75015 Paris, France
- Université Paris Cité, 75014 Paris, France
| | - Irene Pitard
- Structural Bioinformatic Unit, Department of Structural Biology and Chemistry, Institut Pasteur, Université Paris Cité, CNRS UMR 3528, 75015 Paris, France
- Université Paris Sorbonne, 75231 Paris, France
| | - Alexandre Chenal
- Biochemistry of Macromolecular Interactions Unit, Department of Structural Biology and Chemistry, Institut Pasteur, Université Paris Cité, CNRS UMR 3528, 75015 Paris, France
| | - Daniel Ladant
- Biochemistry of Macromolecular Interactions Unit, Department of Structural Biology and Chemistry, Institut Pasteur, Université Paris Cité, CNRS UMR 3528, 75015 Paris, France
- Correspondence: ; Tel.: +33-1-4568-8400
| |
Collapse
|
12
|
Saiyasit N, Butlig EAR, Chaney SD, Traylor MK, Hawley NA, Randall RB, Bobinger HV, Frizell CA, Trimm F, Crook ED, Lin M, Hill BD, Keller JL, Nelson AR. Neurovascular Dysfunction in Diverse Communities With Health Disparities-Contributions to Dementia and Alzheimer's Disease. Front Neurosci 2022; 16:915405. [PMID: 35844216 PMCID: PMC9279126 DOI: 10.3389/fnins.2022.915405] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/31/2022] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease and related dementias (ADRD) are an expanding worldwide crisis. In the absence of scientific breakthroughs, the global prevalence of ADRD will continue to increase as more people are living longer. Racial or ethnic minority groups have an increased risk and incidence of ADRD and have often been neglected by the scientific research community. There is mounting evidence that vascular insults in the brain can initiate a series of biological events leading to neurodegeneration, cognitive impairment, and ADRD. We are a group of researchers interested in developing and expanding ADRD research, with an emphasis on vascular contributions to dementia, to serve our local diverse community. Toward this goal, the primary objective of this review was to investigate and better understand health disparities in Alabama and the contributions of the social determinants of health to those disparities, particularly in the context of vascular dysfunction in ADRD. Here, we explain the neurovascular dysfunction associated with Alzheimer's disease (AD) as well as the intrinsic and extrinsic risk factors contributing to dysfunction of the neurovascular unit (NVU). Next, we ascertain ethnoregional health disparities of individuals living in Alabama, as well as relevant vascular risk factors linked to AD. We also discuss current pharmaceutical and non-pharmaceutical treatment options for neurovascular dysfunction, mild cognitive impairment (MCI) and AD, including relevant studies and ongoing clinical trials. Overall, individuals in Alabama are adversely affected by social and structural determinants of health leading to health disparities, driven by rurality, ethnic minority status, and lower socioeconomic status (SES). In general, these communities have limited access to healthcare and healthy food and other amenities resulting in decreased opportunities for early diagnosis of and pharmaceutical treatments for ADRD. Although this review is focused on the current state of health disparities of ADRD patients in Alabama, future studies must include diversity of race, ethnicity, and region to best be able to treat all individuals affected by ADRD.
Collapse
Affiliation(s)
- Napatsorn Saiyasit
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Evan-Angelo R. Butlig
- Department of Neurology, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA, United States
| | - Samantha D. Chaney
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Miranda K. Traylor
- Department of Health, Kinesiology, and Sport, University of South Alabama, Mobile, AL, United States
| | - Nanako A. Hawley
- Department of Psychology, University of South Alabama, Mobile, AL, United States
| | - Ryleigh B. Randall
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Hanna V. Bobinger
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Carl A. Frizell
- Department of Physician Assistant Studies, University of South Alabama, Mobile, AL, United States
| | - Franklin Trimm
- College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Errol D. Crook
- Department of Internal Medicine, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Mike Lin
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Benjamin D. Hill
- Department of Psychology, University of South Alabama, Mobile, AL, United States
| | - Joshua L. Keller
- Department of Health, Kinesiology, and Sport, University of South Alabama, Mobile, AL, United States
| | - Amy R. Nelson
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| |
Collapse
|
13
|
Teixeira-Nunes M, Retailleau P, Comisso M, Deruelle V, Mechold U, Renault L. Bacterial Nucleotidyl Cyclases Activated by Calmodulin or Actin in Host Cells: Enzyme Specificities and Cytotoxicity Mechanisms Identified to Date. Int J Mol Sci 2022; 23:ijms23126743. [PMID: 35743184 PMCID: PMC9223806 DOI: 10.3390/ijms23126743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 02/06/2023] Open
Abstract
Many pathogens manipulate host cell cAMP signaling pathways to promote their survival and proliferation. Bacterial Exoenzyme Y (ExoY) toxins belong to a family of invasive, structurally-related bacterial nucleotidyl cyclases (NC). Inactive in bacteria, they use proteins that are uniquely and abundantly present in eukaryotic cells to become potent, unregulated NC enzymes in host cells. Other well-known members of the family include Bacillus anthracis Edema Factor (EF) and Bordetella pertussis CyaA. Once bound to their eukaryotic protein cofactor, they can catalyze supra-physiological levels of various cyclic nucleotide monophosphates in infected cells. Originally identified in Pseudomonas aeruginosa, ExoY-related NC toxins appear now to be more widely distributed among various γ- and β-proteobacteria. ExoY-like toxins represent atypical, poorly characterized members within the NC toxin family. While the NC catalytic domains of EF and CyaA toxins use both calmodulin as cofactor, their counterparts in ExoY-like members from pathogens of the genus Pseudomonas or Vibrio use actin as a potent cofactor, in either its monomeric or polymerized form. This is an original subversion of actin for cytoskeleton-targeting toxins. Here, we review recent advances on the different members of the NC toxin family to highlight their common and distinct functional characteristics at the molecular, cytotoxic and enzymatic levels, and important aspects that need further characterizations.
Collapse
Affiliation(s)
- Magda Teixeira-Nunes
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, 91198 Gif-sur-Yvette, France; (M.T.-N.); (M.C.)
| | - Pascal Retailleau
- Institut de Chimie des Substances Naturelles (ICSN), CNRS-UPR2301, Université Paris-Saclay, 1 Avenue de la Terrasse, 91198 Gif-sur-Yvette, France;
| | - Martine Comisso
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, 91198 Gif-sur-Yvette, France; (M.T.-N.); (M.C.)
| | - Vincent Deruelle
- Unité de Biochimie des Interactions Macromoléculaires, Département de Biologie Structurale et Chimie, CNRS UMR 3528, Institut Pasteur, 75015 Paris, France; (V.D.); (U.M.)
| | - Undine Mechold
- Unité de Biochimie des Interactions Macromoléculaires, Département de Biologie Structurale et Chimie, CNRS UMR 3528, Institut Pasteur, 75015 Paris, France; (V.D.); (U.M.)
| | - Louis Renault
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, 91198 Gif-sur-Yvette, France; (M.T.-N.); (M.C.)
- Correspondence:
| |
Collapse
|
14
|
Molecular Mechanisms Involved in Pseudomonas aeruginosa Bacteremia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1386:325-345. [DOI: 10.1007/978-3-031-08491-1_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
|
15
|
Cytotoxic tau released from lung microvascular endothelial cells upon infection with Pseudomonas aeruginosa promotes neuronal tauopathy. J Biol Chem 2021; 298:101482. [PMID: 34896150 PMCID: PMC8718960 DOI: 10.1016/j.jbc.2021.101482] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 11/23/2022] Open
Abstract
Patients who recover from nosocomial pneumonia oftentimes exhibit long-lasting cognitive impairment comparable with what is observed in Alzheimer’s disease patients. We previously hypothesized that the lung endothelium contributes to infection-related neurocognitive dysfunction, because bacteria-exposed endothelial cells release a form(s) of cytotoxic tau that is sufficient to impair long-term potentiation in the hippocampus. However, the full-length lung and endothelial tau isoform(s) have yet to be resolved and it remains unclear whether the infection-induced endothelial cytotoxic tau triggers neuronal tau aggregation. Here, we demonstrate that lung endothelial cells express a big tau isoform and three additional tau isoforms that are similar to neuronal tau, each containing four microtubule-binding repeat domains, and that tau is expressed in lung capillaries in vivo. To test whether infection elicits endothelial tau capable of causing transmissible tau aggregation, the cells were infected with Pseudomonas aeruginosa. The infection-induced tau released from endothelium into the medium-induced neuronal tau aggregation in reporter cells, including reporter cells that express either the four microtubule-binding repeat domains or the full-length tau. Infection-induced release of pathological tau variant(s) from endothelium, and the ability of the endothelial-derived tau to cause neuronal tau aggregation, was abolished in tau knockout cells. After bacterial lung infection, brain homogenates from WT mice, but not from tau knockout mice, initiated tau aggregation. Thus, we conclude that bacterial pneumonia initiates the release of lung endothelial-derived cytotoxic tau, which is capable of propagating a neuronal tauopathy.
Collapse
|
16
|
Karki P, Birukova AA. Microtubules as Major Regulators of Endothelial Function: Implication for Lung Injury. Front Physiol 2021; 12:758313. [PMID: 34777018 PMCID: PMC8582326 DOI: 10.3389/fphys.2021.758313] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 10/06/2021] [Indexed: 01/04/2023] Open
Abstract
Endothelial dysfunction has been attributed as one of the major complications in COVID-19 patients, a global pandemic that has already caused over 4 million deaths worldwide. The dysfunction of endothelial barrier is characterized by an increase in endothelial permeability and inflammatory responses, and has even broader implications in the pathogenesis of acute respiratory syndromes such as ARDS, sepsis and chronic illnesses represented by pulmonary arterial hypertension and interstitial lung disease. The structural integrity of endothelial barrier is maintained by cytoskeleton elements, cell-substrate focal adhesion and adhesive cell junctions. Agonist-mediated changes in endothelial permeability are directly associated with reorganization of actomyosin cytoskeleton leading to cell contraction and opening of intercellular gaps or enhancement of cortical actin cytoskeleton associated with strengthening of endothelial barrier. The role of actin cytoskeleton remodeling in endothelial barrier regulation has taken the central stage, but the impact of microtubules in this process remains less explored and under-appreciated. This review will summarize the current knowledge on the crosstalk between microtubules dynamics and actin cytoskeleton remodeling, describe the signaling mechanisms mediating this crosstalk, discuss epigenetic regulation of microtubules stability and its nexus with endothelial barrier maintenance, and overview a role of microtubules in targeted delivery of signaling molecules regulating endothelial permeability and inflammation.
Collapse
Affiliation(s)
- Pratap Karki
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Anna A Birukova
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
17
|
Belyy A, Merino F, Mechold U, Raunser S. Mechanism of actin-dependent activation of nucleotidyl cyclase toxins from bacterial human pathogens. Nat Commun 2021; 12:6628. [PMID: 34785651 PMCID: PMC8595890 DOI: 10.1038/s41467-021-26889-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 10/25/2021] [Indexed: 12/19/2022] Open
Abstract
Bacterial human pathogens secrete initially inactive nucleotidyl cyclases that become potent enzymes by binding to actin inside eukaryotic host cells. The underlying molecular mechanism of this activation is, however, unclear. Here, we report structures of ExoY from Pseudomonas aeruginosa and Vibrio vulnificus bound to their corresponding activators F-actin and profilin-G-actin. The structures reveal that in contrast to the apo-state, two flexible regions become ordered and interact strongly with actin. The specific stabilization of these regions results in an allosteric stabilization of the nucleotide binding pocket and thereby to an activation of the enzyme. Differences in the sequence and conformation of the actin-binding regions are responsible for the selective binding to either F- or G-actin. Other nucleotidyl cyclase toxins that bind to calmodulin rather than actin undergo a similar disordered-to-ordered transition during activation, suggesting that the allosteric activation-by-stabilization mechanism of ExoY is conserved in these enzymes, albeit the different activator.
Collapse
Affiliation(s)
- Alexander Belyy
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Otto-Hahn-Str. 11, 44227, Dortmund, Germany
| | - Felipe Merino
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Otto-Hahn-Str. 11, 44227, Dortmund, Germany
- Department of Protein Evolution, Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076, Tübingen, Germany
| | - Undine Mechold
- Unité de Biochimie des Interactions Macromoléculaires, Département de Biologie Structurale et Chimie, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Stefan Raunser
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Otto-Hahn-Str. 11, 44227, Dortmund, Germany.
| |
Collapse
|
18
|
Wagener BM, Hu R, Wu S, Pittet JF, Ding Q, Che P. The Role of Pseudomonas aeruginosa Virulence Factors in Cytoskeletal Dysregulation and Lung Barrier Dysfunction. Toxins (Basel) 2021; 13:776. [PMID: 34822560 PMCID: PMC8625199 DOI: 10.3390/toxins13110776] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/24/2021] [Accepted: 10/28/2021] [Indexed: 12/19/2022] Open
Abstract
Pseudomonas (P.) aeruginosa is an opportunistic pathogen that causes serious infections and hospital-acquired pneumonia in immunocompromised patients. P. aeruginosa accounts for up to 20% of all cases of hospital-acquired pneumonia, with an attributable mortality rate of ~30-40%. The poor clinical outcome of P. aeruginosa-induced pneumonia is ascribed to its ability to disrupt lung barrier integrity, leading to the development of lung edema and bacteremia. Airway epithelial and endothelial cells are important architecture blocks that protect the lung from invading pathogens. P. aeruginosa produces a number of virulence factors that can modulate barrier function, directly or indirectly, through exploiting cytoskeleton networks and intercellular junctional complexes in eukaryotic cells. This review summarizes the current knowledge on P. aeruginosa virulence factors, their effects on the regulation of the cytoskeletal network and associated components, and molecular mechanisms regulating barrier function in airway epithelial and endothelial cells. A better understanding of these processes will help to lay the foundation for new therapeutic approaches against P. aeruginosa-induced pneumonia.
Collapse
Affiliation(s)
- Brant M. Wagener
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (B.M.W.); (R.H.); (S.W.); (J.-F.P.); (Q.D.)
- Division of Molecular and Translational Biomedicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Division of Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ruihan Hu
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (B.M.W.); (R.H.); (S.W.); (J.-F.P.); (Q.D.)
- Division of Molecular and Translational Biomedicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Internal Medicine, Guiqian International General Hospital, Guiyang 550024, China
| | - Songwei Wu
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (B.M.W.); (R.H.); (S.W.); (J.-F.P.); (Q.D.)
- Division of Molecular and Translational Biomedicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jean-Francois Pittet
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (B.M.W.); (R.H.); (S.W.); (J.-F.P.); (Q.D.)
- Division of Molecular and Translational Biomedicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Division of Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Qiang Ding
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (B.M.W.); (R.H.); (S.W.); (J.-F.P.); (Q.D.)
- Division of Molecular and Translational Biomedicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Pulin Che
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (B.M.W.); (R.H.); (S.W.); (J.-F.P.); (Q.D.)
- Division of Molecular and Translational Biomedicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
19
|
Silistre H, Raoux-Barbot D, Mancinelli F, Sangouard F, Dupin A, Belyy A, Deruelle V, Renault L, Ladant D, Touqui L, Mechold U. Prevalence of ExoY Activity in Pseudomonas aeruginosa Reference Panel Strains and Impact on Cytotoxicity in Epithelial Cells. Front Microbiol 2021; 12:666097. [PMID: 34675890 PMCID: PMC8524455 DOI: 10.3389/fmicb.2021.666097] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 07/30/2021] [Indexed: 11/13/2022] Open
Abstract
ExoY is among the effectors that are injected by the type III secretion system (T3SS) of Pseudomonas aeruginosa into host cells. Inside eukaryotic cells, ExoY interacts with F-actin, which stimulates its potent nucleotidyl cyclase activity to produce cyclic nucleotide monophosphates (cNMPs). ExoY has broad substrate specificity with GTP as a preferential substrate in vitro. How ExoY contributes to the virulence of P. aeruginosa remains largely unknown. Here, we examined the prevalence of active ExoY among strains from the international P. aeruginosa reference panel, a collection of strains that includes environmental and clinical isolates, commonly used laboratory strains, and sequential clonal isolates from cystic fibrosis (CF) patients and thus represents the large diversity of this bacterial species. The ability to secrete active ExoY was determined by measuring the F-actin stimulated guanylate cyclase (GC) activity in bacterial culture supernatants. We found an overall ExoY activity prevalence of about 60% among the 40 examined strains with no significant difference between CF and non-CF isolates. In parallel, we used cellular infection models of human lung epithelial cells to compare the cytotoxic effects of isogenic reference strains expressing active ExoY or lacking the exoY gene. We found that P. aeruginosa strains lacking ExoY were in fact more cytotoxic to the epithelial cells than those secreting active ExoY. This suggests that under certain conditions, ExoY might partly alleviate the cytotoxic effects of other virulence factors of P. aeruginosa.
Collapse
Affiliation(s)
- Hazel Silistre
- Unité de Biochimie des Interactions Macromoléculaires, Département de Biologie Structurale et Chimie, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Dorothée Raoux-Barbot
- Unité de Biochimie des Interactions Macromoléculaires, Département de Biologie Structurale et Chimie, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Federica Mancinelli
- Unité de Biochimie des Interactions Macromoléculaires, Département de Biologie Structurale et Chimie, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Flora Sangouard
- Unité de Biochimie des Interactions Macromoléculaires, Département de Biologie Structurale et Chimie, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Alice Dupin
- Unité de Biochimie des Interactions Macromoléculaires, Département de Biologie Structurale et Chimie, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Alexander Belyy
- Unité de Biochimie des Interactions Macromoléculaires, Département de Biologie Structurale et Chimie, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Vincent Deruelle
- Unité de Biochimie des Interactions Macromoléculaires, Département de Biologie Structurale et Chimie, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Louis Renault
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Daniel Ladant
- Unité de Biochimie des Interactions Macromoléculaires, Département de Biologie Structurale et Chimie, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Lhousseine Touqui
- Mucoviscidose: Physiopathologie et Phénogénomique, Centre de Recherche Saint-Antoine (CRSA), INSERM UMR S 938, Sorbonne Université, Paris, France.,Mucoviscidose et Bronchopathies Chroniques, Département Santé Globale, Institute Pasteur, Paris, France
| | - Undine Mechold
- Unité de Biochimie des Interactions Macromoléculaires, Département de Biologie Structurale et Chimie, Institut Pasteur, CNRS UMR 3528, Paris, France
| |
Collapse
|
20
|
Balczon R, Lin MT, Lee JY, Abbasi A, Renema P, Voth SB, Zhou C, Koloteva A, Michael Francis C, Sodha NR, Pittet JF, Wagener BM, Bell J, Choi CS, Ventetuolo CE, Stevens T. Pneumonia initiates a tauopathy. FASEB J 2021; 35:e21807. [PMID: 34384141 PMCID: PMC8443149 DOI: 10.1096/fj.202100718r] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/30/2021] [Accepted: 07/06/2021] [Indexed: 12/11/2022]
Abstract
Pneumonia causes short‐ and long‐term cognitive dysfunction in a high proportion of patients, although the mechanism(s) responsible for this effect are unknown. Here, we tested the hypothesis that pneumonia‐elicited cytotoxic amyloid and tau variants: (1) are present in the circulation during infection; (2) lead to impairment of long‐term potentiation; and, (3) inhibit long‐term potentiation dependent upon tau. Cytotoxic amyloid and tau species were recovered from the blood and the hippocampus following pneumonia, and they were present in the extracorporeal membrane oxygenation oxygenators of patients with pneumonia, especially in those who died. Introduction of immunopurified blood‐borne amyloid and tau into either the airways or the blood of uninfected animals acutely and chronically impaired hippocampal information processing. In contrast, the infection did not impair long‐term potentiation in tau knockout mice and the amyloid‐ and tau‐dependent disruption in hippocampal signaling was less severe in tau knockout mice. Moreover, the infection did not elicit cytotoxic amyloid and tau variants in tau knockout mice. Therefore, pneumonia initiates a tauopathy that contributes to cognitive dysfunction.
Collapse
Affiliation(s)
- Ron Balczon
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, USA.,Center for Lung Biology, University of South Alabama, Mobile, AL, USA
| | - Mike T Lin
- Center for Lung Biology, University of South Alabama, Mobile, AL, USA.,Department of Physiology and Cell Biology, College of Medicine, Center for Lung Biology, University of South Alabama, Mobile, AL, USA
| | - Ji Young Lee
- Center for Lung Biology, University of South Alabama, Mobile, AL, USA.,Department of Physiology and Cell Biology, College of Medicine, Center for Lung Biology, University of South Alabama, Mobile, AL, USA.,Internal Medicine, University of South Alabama, Mobile, AL, USA
| | - Adeel Abbasi
- Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Alpert Medical School of Brown University, Providence, RI, USA
| | - Phoibe Renema
- Center for Lung Biology, University of South Alabama, Mobile, AL, USA.,Department of Physiology and Cell Biology, College of Medicine, Center for Lung Biology, University of South Alabama, Mobile, AL, USA
| | - Sarah B Voth
- Center for Lung Biology, University of South Alabama, Mobile, AL, USA.,Department of Physiology and Cell Biology, College of Medicine, Center for Lung Biology, University of South Alabama, Mobile, AL, USA
| | - Chun Zhou
- Center for Lung Biology, University of South Alabama, Mobile, AL, USA.,Department of Physiology and Cell Biology, College of Medicine, Center for Lung Biology, University of South Alabama, Mobile, AL, USA
| | - Anna Koloteva
- Center for Lung Biology, University of South Alabama, Mobile, AL, USA.,Department of Physiology and Cell Biology, College of Medicine, Center for Lung Biology, University of South Alabama, Mobile, AL, USA
| | - C Michael Francis
- Center for Lung Biology, University of South Alabama, Mobile, AL, USA.,Department of Physiology and Cell Biology, College of Medicine, Center for Lung Biology, University of South Alabama, Mobile, AL, USA
| | - Neel R Sodha
- Department of Surgery, Brown University, Providence, RI, USA
| | - Jean-Francois Pittet
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Brant M Wagener
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jessica Bell
- Center for Lung Biology, University of South Alabama, Mobile, AL, USA.,Department of Physiology and Cell Biology, College of Medicine, Center for Lung Biology, University of South Alabama, Mobile, AL, USA
| | - Chung-Sik Choi
- Center for Lung Biology, University of South Alabama, Mobile, AL, USA.,Department of Physiology and Cell Biology, College of Medicine, Center for Lung Biology, University of South Alabama, Mobile, AL, USA
| | - Corey E Ventetuolo
- Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Alpert Medical School of Brown University, Providence, RI, USA.,Health Services, Policy and Practice, Brown University School of Public Health, Providence, RI, USA
| | - Troy Stevens
- Center for Lung Biology, University of South Alabama, Mobile, AL, USA.,Department of Physiology and Cell Biology, College of Medicine, Center for Lung Biology, University of South Alabama, Mobile, AL, USA.,Internal Medicine, University of South Alabama, Mobile, AL, USA
| |
Collapse
|
21
|
Xi J, Ding D, Zhu H, Wang R, Su F, Wu W, Xiao Z, Liang X, Zhao Q, Hong Z, Fu H, Xiao Q. Disturbed microbial ecology in Alzheimer's disease: evidence from the gut microbiota and fecal metabolome. BMC Microbiol 2021; 21:226. [PMID: 34384375 PMCID: PMC8361629 DOI: 10.1186/s12866-021-02286-z] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 07/26/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Gut microbiota (GMB) alteration has been reported to influence the Alzheimer's disease (AD) pathogenesis through immune, endocrine, and metabolic pathways. This study aims to investigate metabolic output of the dysbiosis of GMB in AD pathogenesis. In this study, the fecal microbiota and metabolome from 21 AD participants and 44 cognitively normal control participants were measured. Untargeted GMB taxa was analyzed through 16S ribosomal RNA gene profiling based on next-generation sequencing and fecal metabolites were quantified by using ultrahigh performance liquid chromatography-mass spectrometry (UPLC-MS). RESULTS Our analysis revealed that AD was characterized by 15 altered gut bacterial genera, of which 46.7% (7/15 general) was significantly associated with a series of metabolite markers. The predicted metabolic profile of altered gut microbial composition included steroid hormone biosynthesis, N-Acyl amino acid metabolism and piperidine metabolism. Moreover, a combination of 2 gut bacterial genera (Faecalibacterium and Pseudomonas) and 4 metabolites (N-Docosahexaenoyl GABA, 19-Oxoandrost-4-ene-3,17-dione, Trigofoenoside F and 22-Angeloylbarringtogenol C) was able to discriminate AD from NC with AUC of 0.955 in these 65 subjects. CONCLUSIONS These findings demonstrate that gut microbial alterations and related metabolic output changes may be associated with pathogenesis of AD, and suggest that fecal markers might be used as a non-invasive examination to assist screening and diagnosis of AD.
Collapse
Affiliation(s)
- Jianxiong Xi
- Department of Preventive Medicine and Health Education, School of Public Health, Fudan University, Shanghai, 200032, China
| | - Ding Ding
- Institute of Neurology, Huashan Hospital, Fudan University, Shanghai, 200040, China. .,National Clinical Research Center for Aging Diseases, Shanghai, 200040, China.
| | - Huiwei Zhu
- Department of Preventive Medicine and Health Education, School of Public Health, Fudan University, Shanghai, 200032, China
| | - Ruru Wang
- Department of Preventive Medicine and Health Education, School of Public Health, Fudan University, Shanghai, 200032, China
| | - Feng Su
- Department of Preventive Medicine and Health Education, School of Public Health, Fudan University, Shanghai, 200032, China
| | - Wanqing Wu
- Institute of Neurology, Huashan Hospital, Fudan University, Shanghai, 200040, China.,National Clinical Research Center for Aging Diseases, Shanghai, 200040, China
| | - Zhenxu Xiao
- Institute of Neurology, Huashan Hospital, Fudan University, Shanghai, 200040, China.,National Clinical Research Center for Aging Diseases, Shanghai, 200040, China
| | - Xiaoniu Liang
- Institute of Neurology, Huashan Hospital, Fudan University, Shanghai, 200040, China.,National Clinical Research Center for Aging Diseases, Shanghai, 200040, China
| | - Qianhua Zhao
- Institute of Neurology, Huashan Hospital, Fudan University, Shanghai, 200040, China.,National Clinical Research Center for Aging Diseases, Shanghai, 200040, China
| | - Zhen Hong
- Institute of Neurology, Huashan Hospital, Fudan University, Shanghai, 200040, China.,National Clinical Research Center for Aging Diseases, Shanghai, 200040, China
| | - Hua Fu
- Department of Preventive Medicine and Health Education, School of Public Health, Fudan University, Shanghai, 200032, China
| | - Qianyi Xiao
- Department of Preventive Medicine and Health Education, School of Public Health, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
22
|
cAMP Compartmentalization in Cerebrovascular Endothelial Cells: New Therapeutic Opportunities in Alzheimer's Disease. Cells 2021; 10:cells10081951. [PMID: 34440720 PMCID: PMC8392343 DOI: 10.3390/cells10081951] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/19/2021] [Accepted: 07/28/2021] [Indexed: 12/20/2022] Open
Abstract
The vascular hypothesis used to explain the pathophysiology of Alzheimer’s disease (AD) suggests that a dysfunction of the cerebral microvasculature could be the beginning of alterations that ultimately leads to neuronal damage, and an abnormal increase of the blood–brain barrier (BBB) permeability plays a prominent role in this process. It is generally accepted that, in physiological conditions, cyclic AMP (cAMP) plays a key role in maintaining BBB permeability by regulating the formation of tight junctions between endothelial cells of the brain microvasculature. It is also known that intracellular cAMP signaling is highly compartmentalized into small nanodomains and localized cAMP changes are sufficient at modifying the permeability of the endothelial barrier. This spatial and temporal distribution is maintained by the enzymes involved in cAMP synthesis and degradation, by the location of its effectors, and by the existence of anchor proteins, as well as by buffers or different cytoplasm viscosities and intracellular structures limiting its diffusion. This review compiles current knowledge on the influence of cAMP compartmentalization on the endothelial barrier and, more specifically, on the BBB, laying the foundation for a new therapeutic approach in the treatment of AD.
Collapse
|
23
|
Ding G, Li X, Hou X, Zhou W, Gong Y, Liu F, He Y, Song J, Wang J, Basil P, Li W, Qian S, Saha P, Wang J, Cui C, Yang T, Zou K, Han Y, Amos CI, Xu Y, Chen L, Sun Z. REV-ERB in GABAergic neurons controls diurnal hepatic insulin sensitivity. Nature 2021; 592:763-767. [PMID: 33762728 PMCID: PMC8085086 DOI: 10.1038/s41586-021-03358-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 02/12/2021] [Indexed: 02/08/2023]
Abstract
Systemic insulin sensitivity shows a diurnal rhythm with a peak upon waking1,2. The molecular mechanism that underlies this temporal pattern is unclear. Here we show that the nuclear receptors REV-ERB-α and REV-ERB-β (referred to here as 'REV-ERB') in the GABAergic (γ-aminobutyric acid-producing) neurons in the suprachiasmatic nucleus (SCN) (SCNGABA neurons) control the diurnal rhythm of insulin-mediated suppression of hepatic glucose production in mice, without affecting diurnal eating or locomotor behaviours during regular light-dark cycles. REV-ERB regulates the rhythmic expression of genes that are involved in neurotransmission in the SCN, and modulates the oscillatory firing activity of SCNGABA neurons. Chemogenetic stimulation of SCNGABA neurons at waking leads to glucose intolerance, whereas restoration of the temporal pattern of either SCNGABA neuron firing or REV-ERB expression rescues the time-dependent glucose metabolic phenotype caused by REV-ERB depletion. In individuals with diabetes, an increased level of blood glucose after waking is a defining feature of the 'extended dawn phenomenon'3,4. Patients with type 2 diabetes with the extended dawn phenomenon exhibit a differential temporal pattern of expression of REV-ERB genes compared to patients with type 2 diabetes who do not have the extended dawn phenomenon. These findings provide mechanistic insights into how the central circadian clock regulates the diurnal rhythm of hepatic insulin sensitivity, with implications for our understanding of the extended dawn phenomenon in type 2 diabetes.
Collapse
Affiliation(s)
- Guolian Ding
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA
| | - Xin Li
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA
| | - Xinguo Hou
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China
| | - Wenjun Zhou
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA
| | - Yingyun Gong
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fuqiang Liu
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China
| | - Yanlin He
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Laboratory of Brain Glycemia and Metabolism Control, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Jia Song
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China
| | - Jing Wang
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China
| | - Paul Basil
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA
| | - Wenbo Li
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA
| | - Sichong Qian
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA
| | - Pradip Saha
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Jinbang Wang
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China
| | - Chen Cui
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China
| | - Tingting Yang
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Kexin Zou
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Younghun Han
- Department of Medicine, Section of Epidemiology and Population Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Christopher I Amos
- Department of Medicine, Section of Epidemiology and Population Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Li Chen
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China.
| | - Zheng Sun
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
24
|
Horna G, Ruiz J. Type 3 secretion system of Pseudomonas aeruginosa. Microbiol Res 2021; 246:126719. [PMID: 33582609 DOI: 10.1016/j.micres.2021.126719] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 01/19/2021] [Accepted: 01/26/2021] [Indexed: 12/27/2022]
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen, mainly affecting severe patients, such as those in intensive care units (ICUs). High levels of antibiotic resistance and a long battery of virulence factors characterise this pathogen. Among virulence factors, the T3SS (Type 3 Secretion Systems) are especially relevant, being one of the most important virulence factors in P. aeruginosa. T3SS are a complex "molecular syringe" able to inject different effectors in host cells, subverting cell machinery influencing immune responses, and increasing bacterial survival rates. While T3SS have been largely studied and the molecular structure and main effector functions have been established, a series of questions and further points remain to be clarified or established. The key role of T3SS in P. aeruginosa virulence has resulted in the search for T3SS-targeting molecules able to impair their functions and subsequently improve patient outcomes. This review aims to summarise the most relevant features of the P. aeruginosa T3SS.
Collapse
Affiliation(s)
- Gertrudis Horna
- Universidad Catolica Los Angeles de Chimbote, Instituto de Investigación, Chimbote, Peru.
| | - Joaquim Ruiz
- Laboratorio de Microbiología Molecular y Genómica Bacteriana, Universidad Científica del Sur, Panamericana Sur, Km 19, Lima, Peru.
| |
Collapse
|
25
|
Pittet JF, Hu PJ, Honavar J, Brandon AP, Evans CA, Muthalaly R, Ding Q, Wagener BM. Estrogen Alleviates Sex-Dependent Differences in Lung Bacterial Clearance and Mortality Secondary to Bacterial Pneumonia after Traumatic Brain Injury. J Neurotrauma 2020; 38:989-999. [PMID: 33203297 DOI: 10.1089/neu.2020.7327] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Traumatic brain injury (TBI) is the leading cause of injury-related death and disability in patients under the age of 46 years. Survivors of the initial injury often endure systemic complications such as pulmonary infection, and Pseudomonas aeruginosa is one of the most common causes of nosocomial pneumonia in intensive care units. Female patients are less likely to develop secondary pneumonia after TBI, and pre-clinical studies have revealed a salutary role for estrogen after trauma. Therefore, we hypothesized that female mice would experience less mortality after post-TBI pneumonia with P. aeruginosa. We employed a mouse model of TBI followed by P. aeruginosa pneumonia. Male mice had greater mortality and impaired lung bacterial clearance after post-TBI pneumonia compared with female mice. This was confirmed as a difference in sex hormones, as oophorectomized wild-type mice had mortality and lung bacterial clearance similar to male mice. There were differences in tumor necrosis factor-α secretion in male and female alveolar macrophages after P. aeruginosa infection. Finally, injection of male or oophorectomized wild-type female mice with estrogen restored lung bacterial clearance and prevented mortality. Our model of TBI followed by P. aeruginosa pneumonia is among the first to reveal sex dimorphism in secondary, long-term TBI complications.
Collapse
Affiliation(s)
- Jean-Francois Pittet
- Divisions of Critical Care Medicine and University of Alabama at Birmingham, Birmingham, Alabama, USA.,Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine and University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Parker J Hu
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jaideep Honavar
- Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine and University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Angela P Brandon
- Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine and University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Cilina A Evans
- Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine and University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Rebekah Muthalaly
- Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine and University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Qiang Ding
- Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine and University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Brant M Wagener
- Divisions of Critical Care Medicine and University of Alabama at Birmingham, Birmingham, Alabama, USA.,Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine and University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
26
|
Balczon R, Morrow KA, Leavesley S, Francis CM, Stevens TC, Agwaramgbo E, Williams C, Stevens RP, Langham G, Voth S, Cioffi EA, Weintraub SE, Stevens T. Cystatin C regulates the cytotoxicity of infection-induced endothelial-derived β-amyloid. FEBS Open Bio 2020; 10:2464-2477. [PMID: 33030263 PMCID: PMC7609779 DOI: 10.1002/2211-5463.12997] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 08/25/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023] Open
Abstract
Infection of rat pulmonary microvascular endothelial cells with the bacterium Pseudomonas aeruginosa induces the production and release of cytotoxic oligomeric tau and beta amyloid (Aβ). Here, we characterized these cytotoxic amyloids. Cytotoxic behavior and oligomeric tau were partially resistant to digestion with proteinase K, but cytotoxicity was abolished by various denaturants including phenol, diethylpyrocarbonate (DEPC), and 1,1,1,3,3,3-hexafluoro-2-isopropanol (HFIP). Ultracentrifugation for 8 h at 150 000 g was required to remove cytotoxic activity from the supernatant. Ultracentrifugation, DEPC treatment, and immunodepletion using antibodies against Aβ also demonstrated that cytoprotective protein(s) are released from endothelial cells during P. aeruginosa infection. Mass spectrometry of endothelial cell culture media following P. aeruginosa infection allowed identification of multiple potential secreted modulators of Aβ, including cystatin C, gelsolin, and ApoJ/clusterin. Immunodepletion, co-immunoprecipitation, and ultracentrifugation determined that the cytoprotective factor released during infection of endothelial cells by P. aeruginosa is cystatin C, which appears to be in a complex with Aβ. Cytoprotective cystatin C may provide a novel therapeutic avenue for protection against the long-term consequences of infection with P. aeruginosa.
Collapse
Affiliation(s)
- Ron Balczon
- Department of Biochemistry and Molecular BiologyUniversity of South AlabamaMobileALUSA
- Center for Lung BiologyUniversity of South AlabamaMobileALUSA
| | - Kyle A. Morrow
- Department of Cell Biology and PhysiologyEdward Via College of Osteopathic MedicineMonroeLAUSA
| | - Silas Leavesley
- Center for Lung BiologyUniversity of South AlabamaMobileALUSA
- Department of Chemical and Biomedical EngineeringUniversity of South AlabamaMobileALUSA
| | - Christopher M. Francis
- Center for Lung BiologyUniversity of South AlabamaMobileALUSA
- Department of Physiology and Cell BiologyUniversity of South AlabamaMobileALUSA
| | - Trevor C. Stevens
- Center for Lung BiologyUniversity of South AlabamaMobileALUSA
- Department of Physiology and Cell BiologyUniversity of South AlabamaMobileALUSA
| | - Ezinne Agwaramgbo
- Department of Physiology and Cell BiologyUniversity of South AlabamaMobileALUSA
| | | | - Reece P. Stevens
- Center for Lung BiologyUniversity of South AlabamaMobileALUSA
- Department of Physiology and Cell BiologyUniversity of South AlabamaMobileALUSA
| | - Geri Langham
- Department of Physiology and Cell BiologyUniversity of South AlabamaMobileALUSA
| | - Sarah Voth
- Center for Lung BiologyUniversity of South AlabamaMobileALUSA
- Department of Physiology and Cell BiologyUniversity of South AlabamaMobileALUSA
| | - Eugene A. Cioffi
- Department of PharmacologyUniversity of South AlabamaMobileALUSA
| | - Susan E. Weintraub
- Department of Biochemistry and Structural Biology and Mass Spectrometry LaboratoryUniversity of Texas at San Antonio Health Sciences CenterTXUSA
| | - Troy Stevens
- Center for Lung BiologyUniversity of South AlabamaMobileALUSA
- Department of Physiology and Cell BiologyUniversity of South AlabamaMobileALUSA
| |
Collapse
|
27
|
Wagener BM, Anjum N, Evans C, Brandon A, Honavar J, Creighton J, Traber MG, Stuart RL, Stevens T, Pittet JF. α-Tocopherol Attenuates the Severity of Pseudomonas aeruginosa-induced Pneumonia. Am J Respir Cell Mol Biol 2020; 63:234-243. [PMID: 32243761 DOI: 10.1165/rcmb.2019-0185oc] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Pseudomonas aeruginosa is a lethal pathogen that causes high mortality and morbidity in immunocompromised and critically ill patients. The type III secretion system (T3SS) of P. aeruginosa mediates many of the adverse effects of infection with this pathogen, including increased lung permeability in a Toll-like receptor 4/RhoA/PAI-1 (plasminogen activator inhibitor-1)-dependent manner. α-Tocopherol has antiinflammatory properties that may make it a useful adjunct in treatment of this moribund infection. We measured transendothelial and transepithelial resistance, RhoA and PAI-1 activation, stress fiber formation, P. aeruginosa T3SS exoenzyme (ExoY) intoxication into host cells, and survival in a murine model of pneumonia in the presence of P. aeruginosa and pretreatment with α-tocopherol. We found that α-tocopherol alleviated P. aeruginosa-mediated alveolar endothelial and epithelial paracellular permeability by inhibiting RhoA, in part, via PAI-1 activation, and increased survival in a mouse model of P. aeruginosa pneumonia. Furthermore, we found that α-tocopherol decreased the activation of RhoA and PAI-1 by blocking the injection of T3SS exoenzymes into alveolar epithelial cells. P. aeruginosa is becoming increasingly antibiotic resistant. We provide evidence that α-tocopherol could be a useful therapeutic agent for individuals who are susceptible to infection with P. aeruginosa, such as those who are immunocompromised or critically ill.
Collapse
Affiliation(s)
- Brant M Wagener
- Department of Anesthesiology and Perioperative Medicine.,Center for Free Radical Biology, and
| | - Naseem Anjum
- Department of Anesthesiology and Perioperative Medicine
| | - Cilina Evans
- Department of Anesthesiology and Perioperative Medicine
| | | | | | | | - Maret G Traber
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon
| | | | - Troy Stevens
- Department of Pharmacology and Medicine and the Center for Lung Biology, University of South Alabama, Mobile, Alabama
| | - Jean-Francois Pittet
- Department of Anesthesiology and Perioperative Medicine.,Center for Lung Injury and Repair, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
28
|
Hassan WH, Ibrahim AMK, Shany SAS, Salam HSH. Virulence and resistance determinants in Pseudomonas aeruginosa isolated from pericarditis in diseased broiler chickens in Egypt. J Adv Vet Anim Res 2020; 7:452-463. [PMID: 33005671 PMCID: PMC7521822 DOI: 10.5455/javar.2020.g441] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/09/2020] [Accepted: 05/12/2020] [Indexed: 12/12/2022] Open
Abstract
Objective: This study was performed to probe the antimicrobial resistance and virulence genes profiling in Pseudomonas aeruginosa recovered from the cases of pericarditis in broiler chickens. Materials and methods: The samples (n = 250) collected from the cases of pericarditis in broiler chickens were bacteriologically examined. Antimicrobial susceptibility was tested by disc diffusion technique. The isolates were genotypically studied for the presence of antimicrobial resistance and virulence gene traits. Finally, the nucleotide sequence of representative resistance gene (mexR gene) and virulence genes (toxA and lasI genes) was analyzed. Results: P. aeruginosa was isolated from 45 samples (18%). Antimicrobial susceptibility testing revealed multidrug resistance in most of the recovered P. aeruginosa isolates, whereas colistin and imipenem were the furthermost in vitro-sensitive antibiotics. Antimicrobial resistance genes, such as blaCTX, fox, and mexR, were prevalent in 100%, 80%, and 100% of the isolates, respectively. PCR confirmed virulence genes such as toxA, exoY, lasB, and lasI in 100%, 60%, 80%, and 80% of the isolates, respectively. Nucleotide sequence analysis of representative resistance gene (mexR gene) and virulence genes (toxA and lasI genes) revealed a high correlation between P. aeruginosa recovered from pericarditis in broiler chickens in the present study with PAO1 (reference strain) and with other sequences published on the GenBank representing different localities worldwide. Conclusion: It could be concluded that P. aeruginosa recovered from pericarditis in broiler chickens in the current study is highly virulent bacteria, resisting most of the therapeutic agents which not only bear hazards for poultry industry but also represent a public health concern.
Collapse
Affiliation(s)
- Walid Hamdy Hassan
- Department of Bacteriology, Mycology and Immunology, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt
| | | | | | - Hala Sayed Hassan Salam
- Department of Bacteriology, Mycology and Immunology, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
29
|
Renema P, Kozhukhar N, Pastukh V, Spadafora D, Paudel SS, Tambe DT, Alexeyev M, Frank DW, Stevens T. Exoenzyme Y induces extracellular active caspase-7 accumulation independent from apoptosis: modulation of transmissible cytotoxicity. Am J Physiol Lung Cell Mol Physiol 2020; 319:L380-L390. [PMID: 32579398 DOI: 10.1152/ajplung.00508.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Caspase-3 and -7 are executioner caspases whose enzymatic activity is necessary to complete apoptotic cell death. Here, we questioned whether endothelial cell infection leads to caspase-3/7-mediated cell death. Pulmonary microvascular endothelial cells (PMVECs) were infected with Pseudomonas aeruginosa (PA103). PA103 caused cell swelling with a granular appearance, paralleled by intracellular caspase-3/7 activation and cell death. In contrast, PMVEC infection with ExoY+ (PA103 ΔexoUexoT::Tc pUCPexoY) caused cell rounding, but it did not activate intracellular caspase-3/7 and it did not cause cell death. However, ExoY+ led to a time-dependent accumulation of active caspase-7, but not caspase-3, in the supernatant, independent of apoptosis. To study the function of extracellular caspase-7, caspase-7- and caspase-3-deficient PMVECs were generated using clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 technology. Caspase-7 activity was significantly reduced in supernatants from infected caspase-7-deficient cells but was unchanged in supernatants from infected caspase-3 deficient cells, indicating an uncoupling in the mechanism of activation of these two enzymes. Because ExoY+ leads to the release of heat stable amyloid cytotoxins that are responsible for transmissible cytotoxicity, we next questioned whether caspase-7 contributes to the severity of this process. Supernatants obtained from infected caspase-7-deficient cells displayed significantly reduced transmissible cytotoxicity when compared with supernatants from infected wild-type controls, illustrating an essential role for caspase-7 in promoting the potency of transmissible cytotoxicity. Thus, we report a mechanism whereby ExoY+ infection induces active caspase-7 accumulation in the extracellular space, independent of both caspase-3 and cell death, where it modulates ExoY+-induced transmissible cytotoxicity.
Collapse
Affiliation(s)
- Phoibe Renema
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama.,Center for Lung Biology, University of South Alabama, Mobile, Alabama
| | - Natalya Kozhukhar
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama.,Center for Lung Biology, University of South Alabama, Mobile, Alabama
| | - Viktoriya Pastukh
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama.,Center for Lung Biology, University of South Alabama, Mobile, Alabama
| | | | - Sunita Subedi Paudel
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama.,Center for Lung Biology, University of South Alabama, Mobile, Alabama
| | - Dhananjay T Tambe
- Department of Pharmacology, University of South Alabama, Mobile, Alabama.,Department of Mechanical Engineering, University of South Alabama, Mobile, Alabama.,Center for Lung Biology, University of South Alabama, Mobile, Alabama
| | - Mikhail Alexeyev
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama.,Center for Lung Biology, University of South Alabama, Mobile, Alabama
| | - Dara W Frank
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Troy Stevens
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama.,Department of Internal Medicine, University of South Alabama, Mobile, Alabama.,Center for Lung Biology, University of South Alabama, Mobile, Alabama
| |
Collapse
|
30
|
Lin MT, Balczon R, Pittet JF, Wagener BM, Moser SA, Morrow KA, Voth S, Francis CM, Leavesley S, Bell J, Alvarez DF, Stevens T. Nosocomial Pneumonia Elicits an Endothelial Proteinopathy: Evidence for a Source of Neurotoxic Amyloids in Critically Ill Patients. Am J Respir Crit Care Med 2020; 198:1575-1578. [PMID: 30280916 DOI: 10.1164/rccm.201801-0060le] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Mike T Lin
- University of South Alabama School of MedicineMobile, Alabama
| | - Ron Balczon
- University of South Alabama School of MedicineMobile, Alabama
| | | | - Brant M Wagener
- University of Alabama at Birmingham School of MedicineBirmingham, Alabamaand
| | - Stephen A Moser
- University of Alabama at Birmingham School of MedicineBirmingham, Alabamaand
| | - K Adam Morrow
- Alabama College of Osteopathic MedicineDothan, Alabama
| | - Sarah Voth
- University of South Alabama School of MedicineMobile, Alabama
| | | | - Silas Leavesley
- University of South Alabama School of MedicineMobile, Alabama
| | - Jessica Bell
- University of South Alabama School of MedicineMobile, Alabama
| | - Diego F Alvarez
- University of South Alabama School of MedicineMobile, Alabama
| | - Troy Stevens
- University of South Alabama School of MedicineMobile, Alabama
| |
Collapse
|
31
|
Pneumonia-induced endothelial amyloids reduce dendritic spine density in brain neurons. Sci Rep 2020; 10:9327. [PMID: 32518286 PMCID: PMC7283224 DOI: 10.1038/s41598-020-66321-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 05/14/2020] [Indexed: 12/26/2022] Open
Abstract
Pseudomonas aeruginosa pneumonia elicits endothelial cell release of cytotoxic amyloids that can be recovered from the bronchoalveolar lavage and cerebrospinal fluids of critically ill patients. Introduction of these cytotoxic amyloids into the lateral ventricle impairs learning and memory in mice. However, it is unclear whether the amyloids of lung origin (1) are neurotropic, and (2) cause structural remodeling of hippocampal dendrites. Thus, we used electrophysiological studies in brain slices and structural analysis of post-mortem tissues obtained from animals exposed to endothelium-derived amyloids to assess these issues. The amyloids were administered via three different routes, by intracerebroventricular, intratracheal, and intraperitoneal injections. Synaptic long-term potentiation was abolished following intracerebroventricular amyloid injection. Fluorescence dialysis or Golgi-impregnation labeling showed reduced dendritic spine density and destabilized spines of hippocampal pyramidal neurons 4 weeks after intracerebroventricular amyloid injection. In comparison, endothelial amyloids introduced to the airway caused the most prominent dendritic spine density reduction, yet intraperitoneal injection of these amyloids did not affect spine density. Our findings indicate that infection-elicited lung endothelial amyloids are neurotropic and reduce neuronal dendritic spine density in vivo. Amyloids applied into the trachea may either be disseminated through the circulation and cross the blood-brain barrier to access the brain, initiate feed-forward amyloid transmissibility among cells of the blood-brain barrier or access the brain in other ways. Nevertheless, lung-derived amyloids suppress hippocampal signaling and cause injury to neuronal structure.
Collapse
|
32
|
Exoenzyme Y Contributes to End-Organ Dysfunction Caused by Pseudomonas aeruginosa Pneumonia in Critically Ill Patients: An Exploratory Study. Toxins (Basel) 2020; 12:toxins12060369. [PMID: 32512716 PMCID: PMC7354586 DOI: 10.3390/toxins12060369] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 05/31/2020] [Accepted: 06/01/2020] [Indexed: 12/19/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that causes pneumonia in immunocompromised and intensive care unit (ICU) patients. During host infection, P. aeruginosa upregulates the type III secretion system (T3SS), which is used to intoxicate host cells with exoenzyme (Exo) virulence factors. Of the four known Exo virulence factors (U, S, T and Y), ExoU has been shown in prior studies to associate with high mortality rates. Preclinical studies have shown that ExoY is an important edema factor in lung infection caused by P. aeruginosa, although its importance in clinical isolates of P. aeruginosa is unknown. We hypothesized that expression of ExoY would be highly prevalent in clinical isolates and would significantly contribute to patient morbidity secondary to P. aeruginosa pneumonia. A single-center, prospective observational study was conducted at the University of Alabama at Birmingham Hospital. Mechanically ventilated ICU patients with a bronchoalveolar lavage fluid culture positive for P. aeruginosa were included. Enrolled patients were followed from ICU admission to discharge and clinical P. aeruginosa isolates were genotyped for the presence of exoenzyme genes. Ninety-nine patients were enrolled in the study. ExoY was present in 93% of P. aeruginosa clinical isolates. Moreover, ExoY alone (ExoY+/ExoU−) was present in 75% of P. aeruginosa isolates, compared to 2% ExoU alone (ExoY−/ExoU+). We found that bacteria isolated from human samples expressed active ExoY and ExoU, and the presence of ExoY in clinical isolates was associated with end-organ dysfunction. This is the first study we are aware of that demonstrates that ExoY is important in clinical outcomes secondary to nosocomial pneumonia.
Collapse
|
33
|
Voth S, Gwin M, Francis CM, Balczon R, Frank DW, Pittet JF, Wagener BM, Moser SA, Alexeyev M, Housley N, Audia JP, Piechocki S, Madera K, Simmons A, Crawford M, Stevens T. Virulent Pseudomonas aeruginosa infection converts antimicrobial amyloids into cytotoxic prions. FASEB J 2020; 34:9156-9179. [PMID: 32413239 PMCID: PMC7383673 DOI: 10.1096/fj.202000051rrr] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 04/19/2020] [Accepted: 04/21/2020] [Indexed: 01/05/2023]
Abstract
Pseudomonas aeruginosa infection elicits the production of cytotoxic amyloids from lung endothelium, yet molecular mechanisms of host‐pathogen interaction that underlie the amyloid production are not well understood. We examined the importance of type III secretion system (T3SS) effectors in the production of cytotoxic amyloids. P aeruginosa possessing a functional T3SS and effectors induced the production and release of cytotoxic amyloids from lung endothelium, including beta amyloid, and tau. T3SS effector intoxication was sufficient to generate cytotoxic amyloid release, yet intoxication with exoenzyme Y (ExoY) alone or together with exoenzymes S and T (ExoS/T/Y) generated the most virulent amyloids. Infection with lab and clinical strains engendered cytotoxic amyloids that were capable of being propagated in endothelial cell culture and passed to naïve cells, indicative of a prion strain. Conversely, T3SS‐incompetent P aeruginosa infection produced non‐cytotoxic amyloids with antimicrobial properties. These findings provide evidence that (1) endothelial intoxication with ExoY is sufficient to elicit self‐propagating amyloid cytotoxins during infection, (2) pulmonary endothelium contributes to innate immunity by generating antimicrobial amyloids in response to bacterial infection, and (3) ExoY contributes to the virulence arsenal of P aeruginosa through the subversion of endothelial amyloid host‐defense to promote a lung endothelial‐derived cytotoxic proteinopathy.
Collapse
Affiliation(s)
- Sarah Voth
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, USA.,Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, AL, USA
| | - Meredith Gwin
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, USA.,Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, AL, USA
| | - Christopher Michael Francis
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, USA.,Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, AL, USA
| | - Ron Balczon
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, AL, USA.,Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL, USA
| | - Dara W Frank
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jean-Francois Pittet
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| | - Brant M Wagener
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| | - Stephen A Moser
- Department of Pathology, University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| | - Mikhail Alexeyev
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, USA.,Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, AL, USA
| | - Nicole Housley
- Department of Microbiology and Immunology, College of Medicine, University of South Alabama, Mobile, AL, USA
| | - Jonathon P Audia
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, AL, USA.,Department of Microbiology and Immunology, College of Medicine, University of South Alabama, Mobile, AL, USA
| | - Scott Piechocki
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, USA
| | - Kayla Madera
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, USA
| | - Autumn Simmons
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, USA
| | - Michaela Crawford
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, USA
| | - Troy Stevens
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, USA.,Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, AL, USA.,Department of Internal Medicine, College of Medicine, University of South Alabama, Mobile, AL, USA
| |
Collapse
|
34
|
Lucas R, Hadizamani Y, Gonzales J, Gorshkov B, Bodmer T, Berthiaume Y, Moehrlen U, Lode H, Huwer H, Hudel M, Mraheil MA, Toque HAF, Chakraborty T, Hamacher J. Impact of Bacterial Toxins in the Lungs. Toxins (Basel) 2020; 12:toxins12040223. [PMID: 32252376 PMCID: PMC7232160 DOI: 10.3390/toxins12040223] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/30/2020] [Accepted: 03/31/2020] [Indexed: 12/13/2022] Open
Abstract
Bacterial toxins play a key role in the pathogenesis of lung disease. Based on their structural and functional properties, they employ various strategies to modulate lung barrier function and to impair host defense in order to promote infection. Although in general, these toxins target common cellular signaling pathways and host compartments, toxin- and cell-specific effects have also been reported. Toxins can affect resident pulmonary cells involved in alveolar fluid clearance (AFC) and barrier function through impairing vectorial Na+ transport and through cytoskeletal collapse, as such, destroying cell-cell adhesions. The resulting loss of alveolar-capillary barrier integrity and fluid clearance capacity will induce capillary leak and foster edema formation, which will in turn impair gas exchange and endanger the survival of the host. Toxins modulate or neutralize protective host cell mechanisms of both the innate and adaptive immunity response during chronic infection. In particular, toxins can either recruit or kill central players of the lung's innate immune responses to pathogenic attacks, i.e., alveolar macrophages (AMs) and neutrophils. Pulmonary disorders resulting from these toxin actions include, e.g., acute lung injury (ALI), the acute respiratory syndrome (ARDS), and severe pneumonia. When acute infection converts to persistence, i.e., colonization and chronic infection, lung diseases, such as bronchitis, chronic obstructive pulmonary disease (COPD), and cystic fibrosis (CF) can arise. The aim of this review is to discuss the impact of bacterial toxins in the lungs and the resulting outcomes for pathogenesis, their roles in promoting bacterial dissemination, and bacterial survival in disease progression.
Collapse
Affiliation(s)
- Rudolf Lucas
- Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA;
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA;
- Department of Medicine and Division of Pulmonary Critical Care Medicine, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA;
- Correspondence: (R.L.); (J.H.); Tel.: +41-31-300-35-00 (J.H.)
| | - Yalda Hadizamani
- Lungen-und Atmungsstiftung, Bern, 3012 Bern, Switzerland;
- Pneumology, Clinic for General Internal Medicine, Lindenhofspital Bern, 3012 Bern, Switzerland
| | - Joyce Gonzales
- Department of Medicine and Division of Pulmonary Critical Care Medicine, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA;
| | - Boris Gorshkov
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA;
| | - Thomas Bodmer
- Labormedizinisches Zentrum Dr. Risch, Waldeggstr. 37 CH-3097 Liebefeld, Switzerland;
| | - Yves Berthiaume
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada;
| | - Ueli Moehrlen
- Pediatric Surgery, University Children’s Hospital, Zürich, Steinwiesstrasse 75, CH-8032 Zürch, Switzerland;
| | - Hartmut Lode
- Insitut für klinische Pharmakologie, Charité, Universitätsklinikum Berlin, Reichsstrasse 2, D-14052 Berlin, Germany;
| | - Hanno Huwer
- Department of Cardiothoracic Surgery, Voelklingen Heart Center, 66333 Voelklingen/Saar, Germany;
| | - Martina Hudel
- Justus-Liebig-University, Biomedical Research Centre Seltersberg, Schubertstr. 81, 35392 Giessen, Germany; (M.H.); (M.A.M.); (T.C.)
| | - Mobarak Abu Mraheil
- Justus-Liebig-University, Biomedical Research Centre Seltersberg, Schubertstr. 81, 35392 Giessen, Germany; (M.H.); (M.A.M.); (T.C.)
| | - Haroldo Alfredo Flores Toque
- Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA;
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA;
| | - Trinad Chakraborty
- Justus-Liebig-University, Biomedical Research Centre Seltersberg, Schubertstr. 81, 35392 Giessen, Germany; (M.H.); (M.A.M.); (T.C.)
| | - Jürg Hamacher
- Lungen-und Atmungsstiftung, Bern, 3012 Bern, Switzerland;
- Pneumology, Clinic for General Internal Medicine, Lindenhofspital Bern, 3012 Bern, Switzerland
- Medical Clinic V-Pneumology, Allergology, Intensive Care Medicine and Environmental Medicine, Faculty of Medicine, Saarland University, University Medical Centre of the Saarland, D-66421 Homburg, Germany
- Institute for Clinical & Experimental Surgery, Faculty of Medicine, Saarland University, D-66421 Homburg, Germany
- Correspondence: (R.L.); (J.H.); Tel.: +41-31-300-35-00 (J.H.)
| |
Collapse
|
35
|
Haldar B, Hamilton CL, Solodushko V, Abney KA, Alexeyev M, Honkanen RE, Scammell JG, Cioffi DL. S100A6 is a positive regulator of PPP5C-FKBP51-dependent regulation of endothelial calcium signaling. FASEB J 2020; 34:3179-3196. [PMID: 31916625 DOI: 10.1096/fj.201901777r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 10/18/2019] [Accepted: 12/19/2019] [Indexed: 11/11/2022]
Abstract
ISOC is a cation current permeating the ISOC channel. In pulmonary endothelial cells, ISOC activation leads to formation of inter-endothelial cell gaps and barrier disruption. The immunophilin FK506-binding protein 51 (FKBP51), in conjunction with the serine/threonine protein phosphatase 5C (PPP5C), inhibits ISOC . Free PPP5C assumes an autoinhibitory state, which has low "basal" catalytic activity. Several S100 protein family members bind PPP5C increasing PPP5C catalytic activity in vitro. One of these family members, S100A6, exhibits a calcium-dependent translocation to the plasma membrane. The goal of this study was to determine whether S100A6 activates PPP5C in pulmonary endothelial cells and contributes to ISOC inhibition by the PPP5C-FKBP51 axis. We observed that S100A6 activates PPP5C to dephosphorylate tau T231. Following ISOC activation, cytosolic S100A6 translocates to the plasma membrane and interacts with the TRPC4 subunit of the ISOC channel. Global calcium entry and ISOC are decreased by S100A6 in a PPP5C-dependent manner and by FKBP51 in a S100A6-dependent manner. Further, calcium entry-induced endothelial barrier disruption is decreased by S100A6 dependent upon PPP5C, and by FKBP51 dependent upon S100A6. Overall, these data reveal that S100A6 plays a key role in the PPP5C-FKBP51 axis to inhibit ISOC and protect the endothelial barrier against calcium entry-induced disruption.
Collapse
Affiliation(s)
- Barnita Haldar
- Departments of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, USA.,Center for Lung Biology, University of South Alabama, Mobile, AL, USA
| | - Caleb L Hamilton
- Department of Anatomy and Molecular Medicine, Alabama College of Osteopathic Medicine, Dothan, AL, USA
| | - Viktoriya Solodushko
- Departments of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, USA
| | - Kevin A Abney
- Departments of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, USA
| | - Mikhail Alexeyev
- Center for Lung Biology, University of South Alabama, Mobile, AL, USA.,Physiology and Cell Biology, University of South Alabama, Mobile, AL, USA
| | - Richard E Honkanen
- Departments of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, USA
| | | | - Donna L Cioffi
- Departments of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, USA.,Center for Lung Biology, University of South Alabama, Mobile, AL, USA
| |
Collapse
|
36
|
Che P, Wagener BM, Zhao X, Brandon AP, Evans CA, Cai GQ, Zhao R, Xu ZX, Han X, Pittet JF, Ding Q. Neuronal Wiskott-Aldrich syndrome protein regulates Pseudomonas aeruginosa-induced lung vascular permeability through the modulation of actin cytoskeletal dynamics. FASEB J 2020; 34:3305-3317. [PMID: 31916311 DOI: 10.1096/fj.201902915r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/23/2019] [Accepted: 12/24/2019] [Indexed: 02/06/2023]
Abstract
Pulmonary edema associated with increased vascular permeability is a severe complication of Pseudomonas (P.) aeruginosa-induced acute lung injury. The mechanisms underlying P aeruginosa-induced vascular permeability are not well understood. In the present study, we investigated the role of neuronal Wiskott Aldrich syndrome protein (N-WASP) in modulating P aeruginosa-induced vascular permeability. Using lung microvascular endothelial and alveolar epithelial cells, we demonstrated that N-WASP downregulation attenuated P aeruginosa-induced actin stress fiber formation and prevented paracellular permeability. P aeruginosa-induced dissociation between VE-cadherin and β-catenin, but increased association between N-WASP and VE-cadherin, suggesting a role for N-WASP in promoting P aeruginosa-induced adherens junction rupture. P aeruginosa increased N-WASP-Y256 phosphorylation, which required the activation of Rho GTPase and focal adhesion kinase. Increased N-WASP-Y256 phosphorylation promotes N-WASP and integrin αVβ6 association as well as TGF-β-mediated permeability across alveolar epithelial cells. Inhibition of N-WASP-Y256 phosphorylation by N-WASP-Y256F overexpression blocked N-WASP effects in P aeruginosa-induced actin stress fiber formation and increased paracellular permeability. In vivo, N-WASP knockdown attenuated the development of pulmonary edema and improved survival in a mouse model of P aeruginosa pneumonia. Together, our data demonstrate that N-WASP plays an essential role in P aeruginosa-induced vascular permeability and pulmonary edema through the modulation of actin cytoskeleton dynamics.
Collapse
Affiliation(s)
- Pulin Che
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.,Molecular and Translational Biomedicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Brant M Wagener
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.,Molecular and Translational Biomedicine, University of Alabama at Birmingham, Birmingham, AL, USA.,Divisions of Critical Care, University of Alabama at Birmingham, Birmingham, AL, USA.,Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Xueke Zhao
- Department of Infectious Diseases, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Angela P Brandon
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Cilina A Evans
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Guo-Qiang Cai
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rui Zhao
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Zhi-Xiang Xu
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Xiaosi Han
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jean-Francois Pittet
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.,Divisions of Critical Care, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Qiang Ding
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.,Molecular and Translational Biomedicine, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
37
|
Balczon R, Pittet JF, Wagener BM, Moser SA, Voth S, Vorhees CV, Williams MT, Bridges JP, Alvarez DF, Koloteva A, Xu Y, Zha XM, Audia JP, Stevens T, Lin MT. Infection-induced endothelial amyloids impair memory. FASEB J 2019; 33:10300-10314. [PMID: 31211919 PMCID: PMC6704457 DOI: 10.1096/fj.201900322r] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 05/21/2019] [Indexed: 01/14/2023]
Abstract
Patients with nosocomial pneumonia exhibit elevated levels of neurotoxic amyloid and tau proteins in the cerebrospinal fluid (CSF). In vitro studies indicate that pulmonary endothelium infected with clinical isolates of either Pseudomonas aeruginosa, Klebsiella pneumoniae, or Staphylococcus aureus produces and releases cytotoxic amyloid and tau proteins. However, the effects of the pulmonary endothelium-derived amyloid and tau proteins on brain function have not been elucidated. Here, we show that P. aeruginosa infection elicits accumulation of detergent insoluble tau protein in the mouse brain and inhibits synaptic plasticity. Mice receiving endothelium-derived amyloid and tau proteins via intracerebroventricular injection exhibit a learning and memory deficit in object recognition, fear conditioning, and Morris water maze studies. We compared endothelial supernatants obtained after the endothelia were infected with P. aeruginosa possessing an intact [P. aeruginosa isolated from patient 103 (PA103) supernatant] or defective [mutant strain of P. aeruginosa lacking a functional type 3 secretion system needle tip complex (ΔPcrV) supernatant] type 3 secretion system. Whereas the PA103 supernatant impaired working memory, the ΔPcrV supernatant had no effect. Immunodepleting amyloid or tau proteins from the PA103 supernatant with the A11 or T22 antibodies, respectively, overtly rescued working memory. Recordings from hippocampal slices treated with endothelial supernatants or CSF from patients with or without nosocomial pneumonia indicated that endothelium-derived neurotoxins disrupted the postsynaptic synaptic response. Taken together, these results establish a plausible mechanism for the neurologic sequelae consequent to nosocomial bacterial pneumonia.-Balczon, R., Pittet, J.-F., Wagener, B. M., Moser, S. A., Voth, S., Vorhees, C. V., Williams, M. T., Bridges, J. P., Alvarez, D. F., Koloteva, A., Xu, Y., Zha, X.-M., Audia, J. P., Stevens, T., Lin, M. T. Infection-induced endothelial amyloids impair memory.
Collapse
Affiliation(s)
- Ron Balczon
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, USA
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA
| | - Jean-Francois Pittet
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, USA
| | - Brant M. Wagener
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, USA
| | - Stephen A. Moser
- Department of Pathology, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, USA
| | - Sarah Voth
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, USA
| | - Charles V. Vorhees
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
- Department of Environmental Health, University of Cincinnati, Cincinnati, Ohio, USA
| | | | - James P. Bridges
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Diego F. Alvarez
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, USA
| | - Anna Koloteva
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, USA
| | - Yuanyuan Xu
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, USA
| | - Xiang-Ming Zha
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, USA
| | - Jonathon P. Audia
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA
- Department of Microbiology and Immunology, University of South Alabama, Mobile, Alabama, USA
| | - Troy Stevens
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, USA
- Department of Internal Medicine, University of South Alabama, Mobile, Alabama, USA
| | - Mike T. Lin
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, USA
| |
Collapse
|
38
|
Lipid Droplets: A Significant but Understudied Contributor of Host⁻Bacterial Interactions. Cells 2019; 8:cells8040354. [PMID: 30991653 PMCID: PMC6523240 DOI: 10.3390/cells8040354] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 04/05/2019] [Accepted: 04/12/2019] [Indexed: 12/13/2022] Open
Abstract
Lipid droplets (LDs) are cytosolic lipid storage organelles that are important for cellular lipid metabolism, energy homeostasis, cell signaling, and inflammation. Several bacterial, viral and protozoal pathogens exploit host LDs to promote infection, thus emphasizing the importance of LDs at the host–pathogen interface. In this review, we discuss the thus far reported relation between host LDs and bacterial pathogens including obligate and facultative intracellular bacteria, and extracellular bacteria. Although there is less evidence for a LD–extracellular bacterial interaction compared to interactions with intracellular bacteria, in this review, we attempt to compare the bacterial mechanisms that target LDs, the host signaling pathways involved and the utilization of LDs by these bacteria. Many intracellular bacteria employ unique mechanisms to target host LDs and potentially obtain nutrients and lipids for vacuolar biogenesis and/or immune evasion. However, extracellular bacteria utilize LDs to either promote host tissue damage or induce host death. We also identify several areas that require further investigation. Along with identifying LD interactions with bacteria besides the ones reported, the precise mechanisms of LD targeting and how LDs benefit pathogens should be explored for the bacteria discussed in the review. Elucidating LD–bacterial interactions promises critical insight into a novel host–pathogen interaction.
Collapse
|
39
|
Emaneini M, Kalantar-Neyestanaki D, Jabalameli L, Hashemi M, Beigverdi R, Jabalameli F. Molecular analysis and antimicrobial resistance pattern of distinct strains of Pseudomonas aeruginosa isolated from cystic fibrosis patients in Iran. IRANIAN JOURNAL OF MICROBIOLOGY 2019; 11:98-107. [PMID: 31341563 PMCID: PMC6635311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND AND OBJECTIVES Colonization of Pseudomonas aeruginosa in Cystic Fibrosis (CF) patients may lead to severe pulmonary disease and death. Different characteristics of P. aeruginosa from these patients were determined in the present study. MATERIALS AND METHODS Antimicrobial susceptibility and AmpC-overproduction were determined. The β-lactamase genes were detected by PCR and the oprD gene was sequenced in some of the carbapenem resistance isolates. Distribution of exo genes was determined by PCR. Cytotoxicity of Exo effector proteins was measured using A549 cells. Biofilm production was determined by microtiter plate assay. Random amplified polymorphic DNA (RAPD) -PCR was performed for molecular analysis. RESULTS Polymyxin B, piperacillin/tazobactam and meropenem were the most active antibiotics and 9.6% of isolates were ampC overproducers. The prevalence of bla VEB, bla OXA, bla VIM, and bla PER genes were as follow: 22.7%, 3.75%, 6.25% and 3.75%, respectively. A high proportion (83.5%) of isolates was able to produce biofilm. The exoT gene was present in all isolates while exoU was present in about 35% of them. RAPD-PCR revealed 49 patterns among 78 tested isolates in which 34 patterns were detected once. CONCLUSION Biofilm formation ability and relatively high frequency of exoS may contribute to the persistence of bacteria within lungs of CF patients. Some characteristics of isolates recovered from a single patient after several sampling procedures were similar, while others lacked resemblance.
Collapse
Affiliation(s)
- Mohammad Emaneini
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Leila Jabalameli
- Department of Microbiology, Karaj Branch, Islamic Azad University, Karaj, Iran
| | - Mojtaba Hashemi
- Department of Pediatric Gastroenterology, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Reza Beigverdi
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fereshteh Jabalameli
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran,Corresponding author: Fereshteh Jabalameli, Ph.D, Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran. Tel- Fax: +98-021-88955810,
| |
Collapse
|
40
|
Chua MD, Liou CH, Bogdan AC, Law HT, Yeh KM, Lin JC, Siu LK, Guttman JA. Klebsiella pneumoniae disassembles host microtubules in lung epithelial cells. Cell Microbiol 2018; 21:e12977. [PMID: 30415487 DOI: 10.1111/cmi.12977] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 10/29/2018] [Accepted: 10/31/2018] [Indexed: 01/21/2023]
Abstract
Klebsiella pneumoniae raises significant concerns to the health care industry as these microbes are the source of widespread contamination of medical equipment, cause pneumonia as well as other multiorgan metastatic infections and have gained multidrug resistance. Despite soaring mortality rates, the host cell alterations occurring during these infections remain poorly understood. Here, we show that during in vitro and in vivo K. pneumoniae infections of lung epithelia, microtubules are severed and then eliminated. This destruction does not require direct association of K. pneumoniae with the host cells, as microtubules are disassembled in cells that are distant from the infecting bacteria. This microtubule dismantling is dependent on the K. pneumoniae (Kp) gene ytfL as non-pathogenic Escherichia coli expressing Kp ytfL disassemble microtubules in the absence of K. pneumoniae itself. Our data points to the host katanin catalytic subunit A like 1 protein (KATNAL1) and the katanin regulatory subunit B1 protein (KATNB1) as the gatekeepers to the microtubule severing event as both proteins localise specifically to microtubule cut sites. Infected cells that had either of these proteins knocked out maintained intact microtubules. Taken together, we have identified a novel mechanism that a bacterial pathogen has exploited to cause microtubule destruction within the host epithelia.
Collapse
Affiliation(s)
- Michael Dominic Chua
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Ci-Hong Liou
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | | | - Hong T Law
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Kuo-Ming Yeh
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Jung-Chung Lin
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - L Kristopher Siu
- Division of Infection Diseases, National Health Research Institutes, Miaoli, Taiwan
| | - Julian Andrew Guttman
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| |
Collapse
|
41
|
Differential regulation of actin-activated nucleotidyl cyclase virulence factors by filamentous and globular actin. PLoS One 2018; 13:e0206133. [PMID: 30419035 PMCID: PMC6231621 DOI: 10.1371/journal.pone.0206133] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 10/08/2018] [Indexed: 12/22/2022] Open
Abstract
Several bacterial pathogens produce nucleotidyl cyclase toxins to manipulate eukaryotic host cells. Inside host cells they are activated by endogenous cofactors to produce high levels of cyclic nucleotides (cNMPs). The ExoY toxin from Pseudomonas aeruginosa (PaExoY) and the ExoY-like module (VnExoY) found in the MARTX (Multifunctional-Autoprocessing Repeats-in-ToXin) toxin of Vibrio nigripulchritudo share modest sequence similarity (~38%) but were both recently shown to be activated by actin after their delivery to the eukaryotic host cell. Here, we further characterized the ExoY-like cyclase of V. nigripulchritudo. We show that, in contrast to PaExoY that requires polymerized actin (F-actin) for maximum activation, VnExoY is selectively activated by monomeric actin (G-actin). These two enzymes also display different nucleotide substrate and divalent cation specificities. In vitro in presence of the cation Mg2+, the F-actin activated PaExoY exhibits a promiscuous nucleotidyl cyclase activity with the substrate preference GTP>ATP≥UTP>CTP, while the G-actin activated VnExoY shows a strong preference for ATP as substrate, as it is the case for the well-known calmodulin-activated adenylate cyclase toxins from Bordetella pertussis or Bacillus anthracis. These results suggest that the actin-activated nucleotidyl cyclase virulence factors despite sharing a common activator may actually display a greater variability of biological effects in infected cells than initially anticipated.
Collapse
|
42
|
Balczon R, Francis M, Leavesley S, Stevens T. Methods for Detecting Cytotoxic Amyloids Following Infection of Pulmonary Endothelial Cells by Pseudomonas aeruginosa. J Vis Exp 2018. [PMID: 30059033 DOI: 10.3791/57447] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Patients who survive pneumonia have elevated death rates in the months following hospital discharge. It has been hypothesized that infection of pulmonary tissue during pneumonia results in the production of long-lived cytotoxins that can lead to subsequent end organ failure. We have developed in vitro assays to test the hypothesis that cytotoxins are produced during pulmonary infection. Isolated rat pulmonary endothelial cells and the bacterium Pseudomonas aeruginosa are used as model systems, and the production of cytoxins following infection of the endothelial cells by the bacteria is demonstrated using cell culture followed by direct quantitation using lactate dehydrogenase assays and a novel microscopic method utilizing ImageJ technology. The amyloid nature of these cytotoxins was demonstrated by thioflavin T binding assays and by immunoblotting and immunodepletion using A11 anti-amyloid antibody. Further analyses using immunoblotting demonstrated that oligomeric tau and Aβ were produced and released by endothelial cells following infection by P. aeruginosa. These methods should be readily adaptable to analyses of human clinical samples.
Collapse
Affiliation(s)
- Ron Balczon
- Department of Biochemistry and Molecular Biology, University of South Alabama; Center for Lung Biology, University of South Alabama;
| | - Michael Francis
- Department of Physiology and Cell Biology, University of South Alabama; Center for Lung Biology, University of South Alabama
| | - Silas Leavesley
- Department of Chemical and Biomolecular Engineering, University of South Alabama; Center for Lung Biology, University of South Alabama
| | - Troy Stevens
- Department of Physiology and Cell Biology, University of South Alabama; Center for Lung Biology, University of South Alabama
| |
Collapse
|
43
|
Khanppnavar B, Datta S. Crystal structure and substrate specificity of ExoY, a unique T3SS mediated secreted nucleotidyl cyclase toxin from Pseudomonas aeruginosa. Biochim Biophys Acta Gen Subj 2018; 1862:2090-2103. [PMID: 29859257 DOI: 10.1016/j.bbagen.2018.05.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 05/15/2018] [Accepted: 05/25/2018] [Indexed: 01/03/2023]
Abstract
BACKGROUND The nucleotidyl cyclase toxin ExoY is an important virulence determinant of Pseudomonas aeruginosa that causes severe acute and chronic infections in immune-compromised individuals. Additionally, this unique T3SS effector shows a striking preference for cUMP, a newly identified non-canonical secondary messenger. Thereby, ExoY is also considered as a potential tool to study unexplored cUMP signaling pathways. METHODS The crystal structure of ExoY was determined at 2.2 Å resolutions by in-situ proteolysis assisted crystallization and Rosetta-molecular replacement method. Additionally, isothermal calorimetric (ITC) and molecular dynamic (MD) simulation studies were also carried out to gain molecular insights into its substrate specificity and catalysis. RESULTS AND CONCLUSION ExoY is a partially unfolded protein with higher propensity to form soluble higher-order oligomers. However, with meticulous attempts of removing of disordered regions by proteases, the recalcitrant ExoY could be successfully crystallized. The crystal structure of ExoY revealed similar overall structural fold present in other anthrax toxA family of nucleotidyl cyclases, with two-to-three distinctly conserved regions conferring specificity to eukaryotic binding partner. The in-vitro catalytic preference of ExoY is in the following order: cGMP > cUMP > cAMP > cCMP. The substrate specificity of ExoY mainly depends on its ability to bind NTP in proper geometrical orientations. ExoY also seems to prefer one-metal-ion dependent catalysis than two-metal-ion dependent catalysis. GENERAL SIGNIFICANCE Our results provide much needed structural insight on ExoY, an important virulence determinant of Pseudomonas aeruginosa and an exciting tool to study non-canonical cNMP signaling pathways. ACCESSION NUMBERS The structure factors and coordinate files have been deposited in the Protein Data Bank with accession number 5XNW.
Collapse
Affiliation(s)
- Basavraj Khanppnavar
- Department of Structural Biology and Bioinformatics, CSIR-Indian Institute of Chemical Biology (CSIR-IICB), Kolkata, India; Academy of Scientific and Innovative Research (AcSIR), India
| | - Saumen Datta
- Department of Structural Biology and Bioinformatics, CSIR-Indian Institute of Chemical Biology (CSIR-IICB), Kolkata, India; Academy of Scientific and Innovative Research (AcSIR), India.
| |
Collapse
|
44
|
The Role of Pseudomonas aeruginosa ExoY in an Acute Mouse Lung Infection Model. Toxins (Basel) 2018; 10:toxins10050185. [PMID: 29734720 PMCID: PMC5983241 DOI: 10.3390/toxins10050185] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 04/20/2018] [Accepted: 05/02/2018] [Indexed: 12/31/2022] Open
Abstract
The effector protein Exotoxin Y (ExoY) produced by Pseudomonas aeruginosa is injected via the type III secretion system (T3SS) into host cells. ExoY acts as nucleotidyl cyclase promoting the intracellular accumulation of cyclic nucleotides. To what extent nucleotidyl cyclase activity contributes to the pathogenicity of ExoY and which mechanisms participate in the manifestation of lung infection is still unclear. Here, we used an acute airway infection model in mice to address the role of ExoY in lung infection. In infected lungs, a dose-dependent phenotype of infection with bacteria-expressing ExoY was mirrored by haemorrhage, formation of interstitial oedema in alveolar septa, and infiltration of the perivascular space with erythrocytes and neutrophilic granulocytes. Analyses of the infection process on the cellular and organismal level comparing infections with Pseudomonas aeruginosa mutants expressing either nucleotidyl cyclase-active or -inactive ExoY revealed differential cytokine secretion, increased prevalence of apoptosis, and a break of lung barrier integrity in mice infected with cyclase-active ExoY. Notably, of all measured cyclic nucleotides, only the increase of cyclic UMP in infected mouse lungs coincides temporally with the observed early pathologic changes. In summary, our results suggest that the nucleotidyl cyclase activity of ExoY can contribute to P. aeruginosa acute pathogenicity.
Collapse
|
45
|
Hamilton CL, Abney KA, Vasauskas AA, Alexeyev M, Li N, Honkanen RE, Scammell JG, Cioffi DL. Serine/threonine phosphatase 5 (PP5C/PPP5C) regulates the ISOC channel through a PP5C-FKBP51 axis. Pulm Circ 2017; 8:2045893217753156. [PMID: 29283027 PMCID: PMC6018905 DOI: 10.1177/2045893217753156] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Pulmonary endothelial cells express a store-operated calcium entry current (Isoc), which contributes to inter-endothelial cell gap formation. Isoc is regulated by a heterocomplex of proteins that includes the immunophilin FKBP51. FKBP51 inhibits Isoc by mechanisms that are not fully understood. In pulmonary artery endothelial cells (PAECs) we have shown that FKBP51 increases microtubule polymerization, an event that is critical for Isoc inhibition by FKBP51. In neurons, FKBP51 promotes microtubule stability through facilitation of tau dephosphorylation. However, FKBP51 does not possess phosphatase activity. Protein phosphatase 5 (PP5C/PPP5C) can dephosphorylate tau, and similar to FKBP51, PP5C possesses tetratricopeptide repeats (TPR) that mediate interaction with heat shock protein-90 (HSP90) chaperone/scaffolding complexes. We therefore tested whether PP5C contributes to FKBP51-mediated inhibition of Isoc. Both siRNA-mediated suppression of PP5C expression in PAECs and genetic disruption of PP5C in HEK293 cells attenuate FKBP51-mediated inhibition of Isoc. Reintroduction of catalytically competent, but not catalytically inactive PP5C, restored FKBP51-mediated inhibition of Isoc. PAEC cell fractionation studies identified both PP5C and the ISOC heterocomplex in the same membrane fractions. Further, PP5C co-precipitates with TRPC4, an essential subunit of ISOC channel. Finally, to determine if PP5C is required for FKBP51-mediated inhibition of calcium entry-induced inter-endothelial cell gap formation, we measured gap area by wide-field microscopy and performed biotin gap quantification assay and electric cell-substrate impedance sensing (ECIS®). Collectively, the data presented indicate that suppression of PP5C expression negates the protective effect of FKBP51. These observations identify PP5C as a novel member of the ISOC heterocomplex that is required for FKBP51-mediated inhibition of Isoc.
Collapse
Affiliation(s)
| | | | | | | | - Ni Li
- University of South Alabama
| | | | | | | |
Collapse
|
46
|
Jeon J, Kim YJ, Shin H, Ha UH. T3SS effector ExoY reduces inflammasome-related responses by suppressing bacterial motility and delaying activation of NF-κB and caspase-1. FEBS J 2017; 284:3392-3403. [PMID: 28815941 DOI: 10.1111/febs.14199] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Revised: 07/05/2017] [Accepted: 08/11/2017] [Indexed: 11/28/2022]
Abstract
Type III-secreted effectors are essential for modulating host immune responses during the pathogenesis of Pseudomonas aeruginosa infections. Little is known about the impact of one of the effectors, ExoY, on inflammasome activation, which results in IL-1β production and pyroptotic cell death. In this study, we found that transcriptional expression of Il-1β was induced to a lesser extent in response to an exoY-harboring strain than to a deleted mutant. This suppressive effect of ExoY was verified by complementation assay as well as by direct translocation of exoY into host cells. In addition to the production of IL-1β, pyroptotic cell death was also diminished in response to an exoY-harboring strain. These inflammasome responses were mediated by the adenylate cyclase activity of ExoY, which plays a role in delaying the activation of NF-κB and caspase-1, a key component of inflammasome-mediated responses. Moreover, the negative effects of ExoY on these responses were in part conferred by the suppression of bacterial motility, which could reduce the degree of bacterial contact with cells. Together, these results demonstrate that the adenylate cyclase activity of P. aeruginosa ExoY can reduce inflammasome-related responses by influencing both the host and the bacterium itself by delaying the activation of inflammatory pathways and suppressing bacterial motility.
Collapse
Affiliation(s)
- Jisu Jeon
- Department of Biotechnology and Bioinformatics, Korea University, Sejong, Korea
| | - Yong-Jae Kim
- Department of Biotechnology and Bioinformatics, Korea University, Sejong, Korea
| | - Heesung Shin
- Department of Biotechnology and Bioinformatics, Korea University, Sejong, Korea
| | - Un-Hwan Ha
- Department of Biotechnology and Bioinformatics, Korea University, Sejong, Korea
| |
Collapse
|
47
|
Bacterial Nucleotidyl Cyclase Inhibits the Host Innate Immune Response by Suppressing TAK1 Activation. Infect Immun 2017; 85:IAI.00239-17. [PMID: 28652310 DOI: 10.1128/iai.00239-17] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 06/13/2017] [Indexed: 12/22/2022] Open
Abstract
Exoenzyme Y (ExoY) is a type III secretion system effector found in 90% of the Pseudomonas aeruginosa isolates. Although it is known that ExoY is a soluble nucleotidyl cyclase that increases the cytoplasmic levels of nucleoside 3',5'-cyclic monophosphates (cNMPs) to mediate endothelial Tau phosphorylation and permeability, its functional role in the innate immune response is still poorly understood. Transforming growth factor β-activated kinase 1 (TAK1) is critical for mediating Toll-like receptor (TLR) signaling and subsequent activation of NF-κB and AP-1, which are transcriptional activators of innate immunity. Here, we report that ExoY inhibits proinflammatory cytokine production through suppressing the activation of TAK1 as well as downstream NF-κB and mitogen-activated protein (MAP) kinases. Mice infected with ExoY-deficient P. aeruginosa had higher levels of tumor necrosis factor (TNF) and interleukin-6 (IL-6), more neutrophil recruitment, and a lower bacterial load in lung tissue than mice infected with wild-type P. aeruginosa Taken together, our findings identify a previously unknown mechanism by which P. aeruginosa ExoY inhibits the host innate immune response.
Collapse
|
48
|
Balczon R, Morrow KA, Zhou C, Edmonds B, Alexeyev M, Pittet JF, Wagener BM, Moser SA, Leavesley S, Zha X, Frank DW, Stevens T. Pseudomonas aeruginosa infection liberates transmissible, cytotoxic prion amyloids. FASEB J 2017; 31:2785-2796. [PMID: 28314768 DOI: 10.1096/fj.201601042rr] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 02/26/2017] [Indexed: 12/28/2022]
Abstract
Patients who recover from pneumonia subsequently have elevated rates of death after hospital discharge as a result of secondary organ damage, the causes of which are unknown. We used the bacterium Pseudomonas aeruginosa, a common cause of hospital-acquired pneumonia, as a model for investigating this phenomenon. We show that infection of pulmonary endothelial cells by P. aeruginosa induces production and release of a cytotoxic amyloid molecule with prion characteristics, including resistance to various nucleases and proteases. This cytotoxin was self-propagating, was neutralized by anti-amyloid Abs, and induced death of endothelial cells and neurons. Moreover, the cytotoxin induced edema in isolated lungs. Endothelial cells and isolated lungs were protected from cytotoxin-induced death by stimulation of signal transduction pathways that are linked to prion protein. Analysis of bronchoalveolar lavage fluid collected from human patients with P. aeruginosa pneumonia demonstrated cytotoxic activity, and lavage fluid contained amyloid molecules, including oligomeric τ and Aβ. Demonstration of long-lived cytotoxic agents after Pseudomonas infection may establish a molecular link to the high rates of death as a result of end-organ damage in the months after recovery from pneumonia, and modulation of signal transduction pathways that have been linked to prion protein may provide a mechanism for intervention.-Balczon, R., Morrow, K. A., Zhou, C., Edmonds, B., Alexeyev, M., Pittet, J.-F., Wagener, B. M., Moser, S. A., Leavesley, S., Zha, X., Frank, D. W., Stevens, T. Pseudomonas aeruginosa infection liberates transmissible, cytotoxic prion amyloids.
Collapse
Affiliation(s)
- Ron Balczon
- Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA; .,Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | - K Adam Morrow
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA.,Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | - Chun Zhou
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA.,Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | - Bradley Edmonds
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | - Mikhail Alexeyev
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA.,Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | - Jean-Francois Pittet
- Department of Anesthesia and Perioperative Medicine, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, USA
| | - Brant M Wagener
- Department of Anesthesia and Perioperative Medicine, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, USA
| | - Stephen A Moser
- Department of Pathology, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, USA
| | - Silas Leavesley
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA.,Department of Chemical and Biomedical Engineering, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | - Xiangming Zha
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA.,Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | - Dara W Frank
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Troy Stevens
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA.,Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| |
Collapse
|
49
|
Kopperud RK, Rygh CB, Karlsen TV, Krakstad C, Kleppe R, Hoivik EA, Bakke M, Tenstad O, Selheim F, Lidén Å, Madsen L, Pavlin T, Taxt T, Kristiansen K, Curry FRE, Reed RK, Døskeland SO. Increased microvascular permeability in mice lacking Epac1 (Rapgef3). Acta Physiol (Oxf) 2017; 219:441-452. [PMID: 27096875 PMCID: PMC5073050 DOI: 10.1111/apha.12697] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 03/15/2016] [Accepted: 04/14/2016] [Indexed: 12/22/2022]
Abstract
Aim Maintenance of the blood and extracellular volume requires tight control of endothelial macromolecule permeability, which is regulated by cAMP signalling. This study probes the role of the cAMP mediators rap guanine nucleotide exchange factor 3 and 4 (Epac1 and Epac2) for in vivo control of microvascular macromolecule permeability under basal conditions. Methods Epac1−/− and Epac2−/− C57BL/6J mice were produced and compared with wild‐type mice for transvascular flux of radio‐labelled albumin in skin, adipose tissue, intestine, heart and skeletal muscle. The transvascular leakage was also studied by dynamic contrast‐enhanced magnetic resonance imaging (DCE‐MRI) using the MRI contrast agent Gadomer‐17 as probe. Results Epac1−/− mice had constitutively increased transvascular macromolecule transport, indicating Epac1‐dependent restriction of baseline permeability. In addition, Epac1−/− mice showed little or no enhancement of vascular permeability in response to atrial natriuretic peptide (ANP), whether probed with labelled albumin or Gadomer‐17. Epac2−/− and wild‐type mice had similar basal and ANP‐stimulated clearances. Ultrastructure analysis revealed that Epac1−/− microvascular interendothelial junctions had constitutively less junctional complex. Conclusion Epac1 exerts a tonic inhibition of in vivo basal microvascular permeability. The loss of this tonic action increases baseline permeability, presumably by reducing the interendothelial permeability resistance. Part of the action of ANP to increase permeability in wild‐type microvessels may involve inhibition of the basal Epac1‐dependent activity.
Collapse
Affiliation(s)
- R. K. Kopperud
- Department of Biomedicine; University of Bergen; Bergen Norway
- Centre for Cancer Biomarkers (CCBIO); University of Bergen; Bergen Norway
| | - C. Brekke Rygh
- Department of Biomedicine; University of Bergen; Bergen Norway
| | - T. V. Karlsen
- Department of Biomedicine; University of Bergen; Bergen Norway
| | - C. Krakstad
- Department of Biomedicine; University of Bergen; Bergen Norway
| | - R. Kleppe
- Department of Biomedicine; University of Bergen; Bergen Norway
| | - E. A. Hoivik
- Department of Biomedicine; University of Bergen; Bergen Norway
| | - M. Bakke
- Department of Biomedicine; University of Bergen; Bergen Norway
| | - O. Tenstad
- Department of Biomedicine; University of Bergen; Bergen Norway
| | - F. Selheim
- Department of Biomedicine; University of Bergen; Bergen Norway
| | - Å. Lidén
- Department of Biomedicine; University of Bergen; Bergen Norway
| | - L. Madsen
- Department of Biomedicine; University of Bergen; Bergen Norway
- Department of Biology; University of Copenhagen; Copenhagen Denmark
| | - T. Pavlin
- Department of Biomedicine; University of Bergen; Bergen Norway
| | - T. Taxt
- Department of Biomedicine; University of Bergen; Bergen Norway
| | - K. Kristiansen
- Department of Biology; University of Copenhagen; Copenhagen Denmark
| | - F.-R. E. Curry
- Department of Physiology and Membrane Biology; School of Medicine; University of California; Davis CA USA
| | - R. K. Reed
- Department of Biomedicine; University of Bergen; Bergen Norway
- Centre for Cancer Biomarkers (CCBIO); University of Bergen; Bergen Norway
| | - S. O. Døskeland
- Department of Biomedicine; University of Bergen; Bergen Norway
| |
Collapse
|
50
|
Belyy A, Raoux-Barbot D, Saveanu C, Namane A, Ogryzko V, Worpenberg L, David V, Henriot V, Fellous S, Merrifield C, Assayag E, Ladant D, Renault L, Mechold U. Actin activates Pseudomonas aeruginosa ExoY nucleotidyl cyclase toxin and ExoY-like effector domains from MARTX toxins. Nat Commun 2016; 7:13582. [PMID: 27917880 PMCID: PMC5150216 DOI: 10.1038/ncomms13582] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 10/13/2016] [Indexed: 12/15/2022] Open
Abstract
The nucleotidyl cyclase toxin ExoY is one of the virulence factors injected by the Pseudomonas aeruginosa type III secretion system into host cells. Inside cells, it is activated by an unknown eukaryotic cofactor to synthesize various cyclic nucleotide monophosphates. ExoY-like adenylate cyclases are also found in Multifunctional-Autoprocessing Repeats-in-ToXin (MARTX) toxins produced by various Gram-negative pathogens. Here we demonstrate that filamentous actin (F-actin) is the hitherto unknown cofactor of ExoY. Association with F-actin stimulates ExoY activity more than 10,000 fold in vitro and results in stabilization of actin filaments. ExoY is recruited to actin filaments in transfected cells and alters F-actin turnover. Actin also activates an ExoY-like adenylate cyclase MARTX effector domain from Vibrio nigripulchritudo. Finally, using a yeast genetic screen, we identify actin mutants that no longer activate ExoY. Our results thus reveal a new sub-group within the class II adenylyl cyclase family, namely actin-activated nucleotidyl cyclase (AA-NC) toxins.
Collapse
Affiliation(s)
- Alexander Belyy
- Institut Pasteur, CNRS UMR3528, Unité de Biochimie des Interactions Macromoléculaires, Département de Biologie Structurale et Chimie, 25-28 rue du Docteur Roux, 75724 Paris cedex 15, France
- Department of Bacterial Infections, Gamaleya Research Center, Moscow 123098, Russia
| | - Dorothée Raoux-Barbot
- Institut Pasteur, CNRS UMR3528, Unité de Biochimie des Interactions Macromoléculaires, Département de Biologie Structurale et Chimie, 25-28 rue du Docteur Roux, 75724 Paris cedex 15, France
| | - Cosmin Saveanu
- Institut Pasteur, CNRS UMR3525, Génétique des Interactions Macromoléculaires, Département de Génomes et Génétique, 25-28 rue du Docteur Roux, 75724 Paris cedex 15, France
| | - Abdelkader Namane
- Institut Pasteur, CNRS UMR3525, Génétique des Interactions Macromoléculaires, Département de Génomes et Génétique, 25-28 rue du Docteur Roux, 75724 Paris cedex 15, France
| | - Vasily Ogryzko
- Institut Gustave Roussy, CNRS UMR 8126, Unité de Signaling, Nuclei and Innovations in Oncology, 94805 Villejuif, France
| | - Lina Worpenberg
- Institut Pasteur, CNRS UMR3528, Unité de Biochimie des Interactions Macromoléculaires, Département de Biologie Structurale et Chimie, 25-28 rue du Docteur Roux, 75724 Paris cedex 15, France
| | - Violaine David
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198 Gif-sur-Yvette cedex, France
| | - Veronique Henriot
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198 Gif-sur-Yvette cedex, France
| | - Souad Fellous
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198 Gif-sur-Yvette cedex, France
| | - Christien Merrifield
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198 Gif-sur-Yvette cedex, France
| | - Elodie Assayag
- Institut Pasteur, CNRS UMR3528, Unité de Biochimie des Interactions Macromoléculaires, Département de Biologie Structurale et Chimie, 25-28 rue du Docteur Roux, 75724 Paris cedex 15, France
| | - Daniel Ladant
- Institut Pasteur, CNRS UMR3528, Unité de Biochimie des Interactions Macromoléculaires, Département de Biologie Structurale et Chimie, 25-28 rue du Docteur Roux, 75724 Paris cedex 15, France
| | - Louis Renault
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198 Gif-sur-Yvette cedex, France
| | - Undine Mechold
- Institut Pasteur, CNRS UMR3528, Unité de Biochimie des Interactions Macromoléculaires, Département de Biologie Structurale et Chimie, 25-28 rue du Docteur Roux, 75724 Paris cedex 15, France
| |
Collapse
|