1
|
Qi C, Wang Y, Li X, Zheng C, Gu Y, Hu J, Qiu Y, Xie G, Xu S, Zheng Y, Lv Z, Zheng W. Target inhibition of SPAK in choroid plexus attenuates T cell infiltration and demyelination in experimental autoimmune encephalomyelitis. J Neuroinflammation 2025; 22:80. [PMID: 40082912 PMCID: PMC11907836 DOI: 10.1186/s12974-025-03407-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 03/04/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Disease-modifying therapies (DMTs) that prevent immune cell infiltration into the brain have demonstrated efficacy in multiple sclerosis (MS) treatment. However, their unpredictable adverse effects necessitate the development of safer therapeutic alternatives. The choroid plexus (ChP) functions as a crucial barrier against immune cell invasion, and previous studies have shown that preventing immune cell infiltration across the ChP reduces brain lesion in MS animal models. Understanding ChP barrier regulation is therefore essential for identifying novel therapeutic targets for MS. Here, we explored the role of Ste20-related proline/alanine-rich kinase (SPAK) in experimental autoimmune encephalomyelitis (EAE). METHODS We examined the expression patterns of SPAK signaling in ChP using immunofluorescence in the EAE model. To investigate the roles of SPAK, matrix metalloproteinase (MMP) 2 and MMP9 in EAE pathology, we performed ChP-specific gene manipulation via intracerebroventricular (ICV) injection of recombinant adeno-associated virus 2/5 (rAAV2/5). T cell infiltration into the central nervous system (CNS) was analyzed using CD4 immunostaining and flow cytometry. We employed cell immunofluorescence, transwell assays, and rescue experiments in vitro to study SPAK's effects on ChP epithelial barrier integrity. We also evaluated the protective effects of SPAK-Na-K-2Cl cotransporter-1 (NKCC1) inhibitors (ZT-1a and bumetanide) on immune invasion and demyelination during EAE using pharmacological approaches. RESULTS Following EAE induction, we observed progressive increases in both total and phosphorylated SPAK levels in ChP epithelium. Notably, ChP-specific SPAK knockdown significantly reduced T cell invasion and ameliorated EAE pathology, while SPAK overexpression exacerbated these effects. Bulk RNA sequencing and subsequent qPCR validation revealed that SPAK knockdown decreased the expression of MMP2 and MMP9, MMPs that compromise barrier integrity by degrading tight junction proteins. In vitro studies demonstrated that SPAK overexpression impaired ChP barrier function through the activator protein-1 (AP-1)-MMP2/9-zonula occludens-1 (ZO-1) axis. Furthermore, ChP-specific knockdown of either MMP2 or MMP9 protected against EAE pathology. Additionally, we identified SPAK-NKCC1 antagonists (bumetanide and ZT-1a) as promising therapeutic candidates for MS/EAE treatment. CONCLUSIONS Our findings demonstrate that targeting ChP-SPAK signaling represents a novel therapeutic strategy for MS treatment.
Collapse
Affiliation(s)
- Chenxing Qi
- Department of Neurology, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, 315040, China
| | - Yeping Wang
- Rehabilitation Medicine Center, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Xuhang Li
- Neuroscience Medical Center, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, 315040, China
| | - Cheng Zheng
- Department of Neurology, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, 315040, China
| | - Yi Gu
- Neuroscience Medical Center, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, 315040, China
| | - Junxiao Hu
- Neuroscience Medical Center, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, 315040, China
| | - Yiming Qiu
- Neuroscience Medical Center, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, 315040, China
| | - Guomin Xie
- Department of Neurology, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, 315040, China
| | - Shujun Xu
- Neuroscience Medical Center, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, 315040, China
- Department of Physiology and Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Yuyin Zheng
- Rehabilitation Medicine Center, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China.
| | - Zhongyue Lv
- Department of Neurology, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, 315040, China.
| | - Wu Zheng
- Department of Neurology, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, 315040, China.
- Neuroscience Medical Center, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, 315040, China.
| |
Collapse
|
2
|
DeSpenza T, Kiziltug E, Allington G, Barson DG, McGee S, O'Connor D, Robert SM, Mekbib KY, Nanda P, Greenberg ABW, Singh A, Duy PQ, Mandino F, Zhao S, Lynn A, Reeves BC, Marlier A, Getz SA, Nelson-Williams C, Shimelis H, Walsh LK, Zhang J, Wang W, Prina ML, OuYang A, Abdulkareem AF, Smith H, Shohfi J, Mehta NH, Dennis E, Reduron LR, Hong J, Butler W, Carter BS, Deniz E, Lake EMR, Constable RT, Sahin M, Srivastava S, Winden K, Hoffman EJ, Carlson M, Gunel M, Lifton RP, Alper SL, Jin SC, Crair MC, Moreno-De-Luca A, Luikart BW, Kahle KT. PTEN mutations impair CSF dynamics and cortical networks by dysregulating periventricular neural progenitors. Nat Neurosci 2025; 28:536-557. [PMID: 39994410 PMCID: PMC12038823 DOI: 10.1038/s41593-024-01865-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 12/05/2024] [Indexed: 02/26/2025]
Abstract
Enlargement of the cerebrospinal fluid (CSF)-filled brain ventricles (ventriculomegaly) is a defining feature of congenital hydrocephalus (CH) and an under-recognized concomitant of autism. Here, we show that de novo mutations in the autism risk gene PTEN are among the most frequent monogenic causes of CH and primary ventriculomegaly. Mouse Pten-mutant ventriculomegaly results from aqueductal stenosis due to hyperproliferation of periventricular Nkx2.1+ neural progenitor cells (NPCs) and increased CSF production from hyperplastic choroid plexus. Pten-mutant ventriculomegalic cortices exhibit network dysfunction from increased activity of Nkx2.1+ NPC-derived inhibitory interneurons. Raptor deletion or postnatal everolimus treatment corrects ventriculomegaly, rescues cortical deficits and increases survival by antagonizing mTORC1-dependent Nkx2.1+ NPC pathology. Thus, PTEN mutations concurrently alter CSF dynamics and cortical networks by dysregulating Nkx2.1+ NPCs. These results implicate a nonsurgical treatment for CH, demonstrate a genetic association of ventriculomegaly and ASD, and help explain neurodevelopmental phenotypes refractory to CSF shunting in select individuals with CH.
Collapse
Affiliation(s)
- Tyrone DeSpenza
- Interdepartmental Neuroscience Program, Yale School of Medicine, Yale University, New Haven, CT, USA
- Medical Scientist Training Program, Yale School of Medicine, Yale University, New Haven, CT, USA
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
- Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Emre Kiziltug
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Garrett Allington
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, CT, USA
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons and New York Presbyterian Hospital, New York, NY, USA
| | - Daniel G Barson
- Interdepartmental Neuroscience Program, Yale School of Medicine, Yale University, New Haven, CT, USA
- Medical Scientist Training Program, Yale School of Medicine, Yale University, New Haven, CT, USA
| | | | - David O'Connor
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Stephanie M Robert
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Kedous Y Mekbib
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Pranav Nanda
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ana B W Greenberg
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Amrita Singh
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Phan Q Duy
- Interdepartmental Neuroscience Program, Yale School of Medicine, Yale University, New Haven, CT, USA
- Medical Scientist Training Program, Yale School of Medicine, Yale University, New Haven, CT, USA
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Francesca Mandino
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Shujuan Zhao
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Anna Lynn
- Medical Scientist Training Program, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Benjamin C Reeves
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Arnaud Marlier
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Stephanie A Getz
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Carol Nelson-Williams
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Hermela Shimelis
- Autism & Developmental Medicine Institute, Geisinger, Lewisburg, PA, USA
| | - Lauren K Walsh
- Autism & Developmental Medicine Institute, Geisinger, Lewisburg, PA, USA
| | - Junhui Zhang
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Wei Wang
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Mackenzi L Prina
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Department of Neurobiology, UAB Heersink School of Medicine, Birmingham, AL, USA
| | - Annaliese OuYang
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Asan F Abdulkareem
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Department of Neurobiology, UAB Heersink School of Medicine, Birmingham, AL, USA
| | - Hannah Smith
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - John Shohfi
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Neel H Mehta
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Evan Dennis
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Laetitia R Reduron
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Jennifer Hong
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - William Butler
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Bob S Carter
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Engin Deniz
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Evelyn M R Lake
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - R Todd Constable
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Mustafa Sahin
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Siddharth Srivastava
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kellen Winden
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ellen J Hoffman
- Child Study Center, Yale School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Marina Carlson
- Interdepartmental Neuroscience Program, Yale School of Medicine, Yale University, New Haven, CT, USA
- Child Study Center, Yale School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Murat Gunel
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Richard P Lifton
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY, USA
| | - Seth L Alper
- Division of Nephrology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, and Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Diagnostic Medicine Institute, Geisinger, Danville, PA, USA
| | - Sheng Chih Jin
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael C Crair
- Interdepartmental Neuroscience Program, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Andres Moreno-De-Luca
- Autism & Developmental Medicine Institute, Geisinger, Lewisburg, PA, USA
- Department of Radiology, Diagnostic Medicine Institute, Geisinger, Danville, PA, USA
| | - Bryan W Luikart
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA.
- Department of Neurobiology, UAB Heersink School of Medicine, Birmingham, AL, USA.
| | - Kristopher T Kahle
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA.
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
3
|
Xu H, He J, Du H, Jing X, Liu X. Evaluation of the Choroid Plexus Epithelium Inflammation TLR4/NF-κB/NKCC1 Signal Pathway Activation in the Development of Hydrocephalus. CNS Neurosci Ther 2024; 30:e70085. [PMID: 39450988 PMCID: PMC11503839 DOI: 10.1111/cns.70085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/18/2024] [Accepted: 09/27/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Hydrocephalus is characterized by secretion, circulation, and absorption disorder of cerebrospinal fluid (CSF) with high morbidity and complication rate. The relationship between inflammation and abnormal secretion of CSF by choroid plexus epithelium (CPE) had received more attention. In this study, we aim to detect the role of Toll-like receptor 4/nuclear factor-kappa B/Na+/K+/2Cl-cotransporter 1(TLR4/NF-κB/NKCC1) signal pathway in the development of hydrocephalus. METHOD Hydrocephalus was induced in adult rats (8 weeks) by intracisternal kaolin injection, then pyrrolidinedithiocarbamate (PDTC) and bumetanide were administrated to the rats mode. Then the rat model was evaluated, and ventricular volume was calculated at different time points. Then CPE, cortex, preventricular tissue, and CSF were obtained. Protein expressions of TLR-4, NKCC/serine-threonine STE20/SPS1-related, proline-alanine-rich kinase (SPAK), pNKCC1, pSPAK, GFAP, AQP1, and AQP4 were measured by RT-PCR, western blot, and immunofluorescence (IF) stains in CPE, respectively. RESULT Our data showed that inflammation factors tumor necrosis factor-(TNF-α), interleukin 18(IL-18), and glial fibrillary acidic protein (GFAP) concentrations were significantly higher in the model group than in controls. The TLR4/NF-κB/NKCC1 signal pathway were actived by NF-κB-p65, NKCC1, pNKCC1- pSPAK complex, and Aquaporin1 (AQP1) high expression. PDTC and bumetanide use can help regular TLR4/NF-κB/NKCC1 expression and reduced AQP1 expression by down-regulate NF-B-p65 and inhibiting NKCC1, respectively. As a result, the treatment groups alleviated CPE abnormal secretion and ventricle enlargement. CONCLUSION These results confirmed that the inflammatory reaction contributes TLR4/NF-κB/NKCC1 mediated CPE abnormal secretion and consequent hydrocephalus. Regulation of TLR4/NF-κB/NKCC1 and AQP1 can prevent this process. Our study provides a strong rationale for further exploring alleviating CPE abnormal secretion as a therapeutic perspective of hydrocephalus.
Collapse
Affiliation(s)
- Hao Xu
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhuiChina
| | - Jiawei He
- Department of Neurosurgery, The First Affiliated Hospital of Xiamen UniversitySchool of Medicine, Xiamen UniversityXiamenFujianChina
| | - Hua Du
- Key Laboratory of High Magnetic Field and Ion Beam Physical BiologyHefei Institutes of Physical Science, CASHefeiAnhuiP. R. China
- Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control TechnologyHefei Institutes of Physical Science, CASHefeiAnhuiP. R. China
| | - Xiaolei Jing
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhuiChina
| | - Xinfeng Liu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhuiChina
| |
Collapse
|
4
|
Bhuiyan MIH, Habib K, Sultan MT, Chen F, Jahan I, Weng Z, Rahman MS, Islam R, Foley LM, Hitchens TK, Deng X, Canna SW, Sun D, Cao G. SPAK inhibitor ZT-1a attenuates reactive astrogliosis and oligodendrocyte degeneration in a mouse model of vascular dementia. CNS Neurosci Ther 2024; 30:e14654. [PMID: 38433018 PMCID: PMC10909630 DOI: 10.1111/cns.14654] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 01/08/2024] [Accepted: 01/28/2024] [Indexed: 03/05/2024] Open
Abstract
BACKGROUND Astrogliosis and white matter lesions (WML) are key characteristics of vascular contributions to cognitive impairment and dementia (VCID). However, the molecular mechanisms underlying VCID remain poorly understood. Stimulation of Na-K-Cl cotransport 1 (NKCC1) and its upstream kinases WNK (with no lysine) and SPAK (the STE20/SPS1-related proline/alanine-rich kinase) play a role in astrocytic intracellular Na+ overload, hypertrophy, and swelling. Therefore, in this study, we assessed the effect of SPAK inhibitor ZT-1a on pathogenesis and cognitive function in a mouse model of VCID induced by bilateral carotid artery stenosis (BCAS). METHODS Following sham or BCAS surgery, mice were randomly assigned to receive either vehicle (DMSO) or SPAK inhibitor ZT-1a treatment regimen (days 14-35 post-surgery). Mice were then evaluated for cognitive functions by Morris water maze, WML by ex vivo MRI-DTI analysis, and astrogliosis/demyelination by immunofluorescence and immunoblotting. RESULTS Compared to sham control mice, BCAS-Veh mice exhibited chronic cerebral hypoperfusion and memory impairments, accompanied by significant MRI DTI-detected WML and oligodendrocyte (OL) death. Increased activation of WNK-SPAK-NKCC1-signaling proteins was detected in white matter tissues and in C3d+ GFAP+ cytotoxic astrocytes but not in S100A10+ GFAP+ homeostatic astrocytes in BCAS-Veh mice. In contrast, ZT-1a-treated BCAS mice displayed reduced expression and phosphorylation of NKCC1, decreased astrogliosis, OL death, and WML, along with improved memory functions. CONCLUSION BCAS-induced upregulation of WNK-SPAK-NKCC1 signaling contributes to white matter-reactive astrogliosis, OL death, and memory impairment. Pharmacological inhibition of the SPAK activity has therapeutic potential for alleviating pathogenesis and memory impairment in VCID.
Collapse
Affiliation(s)
- Mohammad Iqbal H. Bhuiyan
- Department of Pharmaceutical Sciences, School of PharmacyUniversity of Texas at El PasoEl PasoTexasUSA
- Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
- Pittsburgh Institute for Neurodegenerative DisordersUniversity of PittsburghPittsburghPennsylvaniaUSA
- Veterans Affairs Pittsburgh Health Care System Pittsburgh Healthcare SystemGeriatric Research Education and Clinical CenterPittsburghPennsylvaniaUSA
| | - Khadija Habib
- Department of Pharmaceutical Sciences, School of PharmacyUniversity of Texas at El PasoEl PasoTexasUSA
| | - Md Tipu Sultan
- Department of Pharmaceutical Sciences, School of PharmacyUniversity of Texas at El PasoEl PasoTexasUSA
| | - Fenghua Chen
- Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Israt Jahan
- Department of Pharmaceutical Sciences, School of PharmacyUniversity of Texas at El PasoEl PasoTexasUSA
- Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Zhongfang Weng
- Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Md Shamim Rahman
- Department of Pharmaceutical Sciences, School of PharmacyUniversity of Texas at El PasoEl PasoTexasUSA
| | | | - Lesley M. Foley
- Animal Imaging CenterUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - T. Kevin Hitchens
- Animal Imaging CenterUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of NeurobiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Xianming Deng
- State Key Laboratory of Cellular Stress Biology, School of Life SciencesXiamen UniversityXiamenFujianChina
| | - Scott W. Canna
- Department of Pediatric RheumatologyThe Children's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Dandan Sun
- Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
- Pittsburgh Institute for Neurodegenerative DisordersUniversity of PittsburghPittsburghPennsylvaniaUSA
- Veterans Affairs Pittsburgh Health Care System Pittsburgh Healthcare SystemGeriatric Research Education and Clinical CenterPittsburghPennsylvaniaUSA
| | - Guodong Cao
- Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
- Veterans Affairs Pittsburgh Health Care System Pittsburgh Healthcare SystemGeriatric Research Education and Clinical CenterPittsburghPennsylvaniaUSA
| |
Collapse
|
5
|
Robert SM, Reeves BC, Kiziltug E, Duy PQ, Karimy JK, Mansuri MS, Marlier A, Allington G, Greenberg ABW, DeSpenza T, Singh AK, Zeng X, Mekbib KY, Kundishora AJ, Nelson-Williams C, Hao LT, Zhang J, Lam TT, Wilson R, Butler WE, Diluna ML, Feinberg P, Schafer DP, Movahedi K, Tannenbaum A, Koundal S, Chen X, Benveniste H, Limbrick DD, Schiff SJ, Carter BS, Gunel M, Simard JM, Lifton RP, Alper SL, Delpire E, Kahle KT. The choroid plexus links innate immunity to CSF dysregulation in hydrocephalus. Cell 2023; 186:764-785.e21. [PMID: 36803604 PMCID: PMC10069664 DOI: 10.1016/j.cell.2023.01.017] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 09/26/2022] [Accepted: 01/12/2023] [Indexed: 02/18/2023]
Abstract
The choroid plexus (ChP) is the blood-cerebrospinal fluid (CSF) barrier and the primary source of CSF. Acquired hydrocephalus, caused by brain infection or hemorrhage, lacks drug treatments due to obscure pathobiology. Our integrated, multi-omic investigation of post-infectious hydrocephalus (PIH) and post-hemorrhagic hydrocephalus (PHH) models revealed that lipopolysaccharide and blood breakdown products trigger highly similar TLR4-dependent immune responses at the ChP-CSF interface. The resulting CSF "cytokine storm", elicited from peripherally derived and border-associated ChP macrophages, causes increased CSF production from ChP epithelial cells via phospho-activation of the TNF-receptor-associated kinase SPAK, which serves as a regulatory scaffold of a multi-ion transporter protein complex. Genetic or pharmacological immunomodulation prevents PIH and PHH by antagonizing SPAK-dependent CSF hypersecretion. These results reveal the ChP as a dynamic, cellularly heterogeneous tissue with highly regulated immune-secretory capacity, expand our understanding of ChP immune-epithelial cell cross talk, and reframe PIH and PHH as related neuroimmune disorders vulnerable to small molecule pharmacotherapy.
Collapse
Affiliation(s)
- Stephanie M Robert
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Benjamin C Reeves
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Emre Kiziltug
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Phan Q Duy
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Jason K Karimy
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - M Shahid Mansuri
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA; Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Arnaud Marlier
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Garrett Allington
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA; Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Ana B W Greenberg
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Tyrone DeSpenza
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Amrita K Singh
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Xue Zeng
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Kedous Y Mekbib
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Adam J Kundishora
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | | | - Le Thi Hao
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Jinwei Zhang
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratory, Exeter EX1 2LU, UK
| | - TuKiet T Lam
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, CT 06520, USA; Keck MS & Proteomics Resource, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Rashaun Wilson
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, CT 06520, USA; Keck MS & Proteomics Resource, Yale University School of Medicine, New Haven, CT 06520, USA
| | - William E Butler
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Michael L Diluna
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Philip Feinberg
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute University of Massachusetts Chan Medical School, Worcester, MA 01655, USA; Medical Scientist Training Program, UMass Chan Medical School, Worcester, MA 01655, USA
| | - Dorothy P Schafer
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Kiavash Movahedi
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1050 Brussels, Belgium; Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, 1050 Brussels, Belgium
| | - Allen Tannenbaum
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY 11794, USA; Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York City, NY 11794, USA
| | - Sunil Koundal
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Xinan Chen
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY 11794, USA
| | - Helene Benveniste
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - David D Limbrick
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Steven J Schiff
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Bob S Carter
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Murat Gunel
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - J Marc Simard
- Department of Neurosurgery, University of Maryland, School of Medicine, Baltimore, MD 21201, USA; Department of Pathology, University of Maryland, School of Medicine, Baltimore, MD 21201, USA; Department of Physiology, University of Maryland, School of Medicine, Baltimore, MD 21201, USA
| | - Richard P Lifton
- Laboratory of Human Genetics and Genomics, the Rockefeller University, New York, NY 10065, USA
| | - Seth L Alper
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Kristopher T Kahle
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA; Department of Neurosurgery and Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
6
|
Jung JU, Jaykumar AB, Cobb MH. WNK1 in Malignant Behaviors: A Potential Target for Cancer? Front Cell Dev Biol 2022; 10:935318. [PMID: 35813203 PMCID: PMC9257110 DOI: 10.3389/fcell.2022.935318] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
Metastasis is the major cause of mortality in cancer patients. Analyses of mouse models and patient data have implicated the protein kinase WNK1 as one of a handful of genes uniquely linked to a subset of invasive cancers. WNK1 signaling pathways are widely implicated in the regulation of ion co-transporters and in controlling cell responses to osmotic stress. In this review we will discuss its actions in tumor malignancy in human cancers and present evidence for its function in invasion, migration, angiogenesis and mesenchymal transition.
Collapse
Affiliation(s)
| | | | - Melanie H. Cobb
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
7
|
Bordagaray MJ, Fernández A, Astorga J, Garrido M, Hernández P, Chaparro A, Lira MJ, Gebicke-Haerter P, Hernández M. CpG Single-Site Methylation Regulates TLR2 Expression in Proinflammatory PBMCs From Apical Periodontitis Individuals. Front Immunol 2022; 13:861665. [PMID: 35300329 PMCID: PMC8921253 DOI: 10.3389/fimmu.2022.861665] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 02/09/2022] [Indexed: 12/19/2022] Open
Abstract
Introduction Apical periodontitis (AP) is a common oral disease caused by the inflammatory destruction of the periapical tissues due to the infection of the root canal system of the tooth. It also contributes to systemic bacterial translocation, where peripheric mononuclear blood cells (PBMCs) can act as carriers. Toll-like receptor (TLR) 2 mediates the response to infection and activates inflammatory responses. DNA methylation can be induced by bacteria and contributes to the modulation of this response. Despite the evidence that supports the participation of PBMCs in immune-inflammatory disorders, the inflammatory profile and epigenetic regulatory mechanisms of PBMCs in AP individuals are unknown. Aim To determine TLR2 gene methylation and inflammatory profiles of PBMCs in AP. Methods Cross-sectional exploratory study. Otherwise, healthy individuals with AP (n=27) and controls (n=30) were included. PMBCs were isolated by a Ficoll gradient, cultured for 24 hours, and both RNA and DNA were extracted. DNA was bisulfite-treated, and specific sites at the promoter region of the TLR2 gene were amplified by qPCR using validated primers. To verify its amplification, agarose gels were performed. Then, the PCR product was sequenced. mRNA expression of TLR2 was determined by qPCR. The soluble levels of 105 inflammatory mediators were first explored with Proteome Profiler Human Cytokine Array Kit. Consequently, tumor necrosis factor (TNF)-α, interleukin (IL)-6, IL-10, IL-6Rα, IL-1β, and IL-12p70 levels were measured by Multiplex assay. Results PBMCs from individuals with AP demonstrated a proinflammatory profile showing higher soluble levels of TNF-α, IL-6, and IL-1β compared to controls (p<0.05). Higher TLR2 expression and higher global methylation pattern of the promoter region of the gene were found in AP compared to controls (p<0.05). The CpGs single-sites at positions -166 and -146 were completely methylated, while the site -102 was totally unmethylated, independently of the presence of AP. DNA methylation of CpG single-sites in positions -77 and +24 was positively associated with TLR2 expression. Conclusions PBMCs from AP subjects show a hyperinflammatory phenotype and TLR2 upregulation in association with single CpG-sites’ methylation from the TLR2 gene promoter, thereby contributing to a sustained systemic inflammatory load in individuals with periapical endodontic diseases.
Collapse
Affiliation(s)
- María José Bordagaray
- Laboratory of Periodontal Biology, Faculty of Dentistry, Universidad de Chile, Santiago, Chile.,Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Alejandra Fernández
- Laboratory of Periodontal Biology, Faculty of Dentistry, Universidad de Chile, Santiago, Chile.,Faculty of Dentistry, Universidad Andres Bello, Santiago, Chile
| | - Jessica Astorga
- Laboratory of Periodontal Biology, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Mauricio Garrido
- Laboratory of Periodontal Biology, Faculty of Dentistry, Universidad de Chile, Santiago, Chile.,Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Patricia Hernández
- Laboratory of Periodontal Biology, Faculty of Dentistry, Universidad de Chile, Santiago, Chile.,Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Alejandra Chaparro
- Department of Periodontology, Faculty of Dentistry, Centro de Investigación e Innovación Biomédica (CIIB), Universidad de Los Andes, Santiago, Chile
| | - María Jesús Lira
- Department of Orthopedic Surgery, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Peter Gebicke-Haerter
- Institute of Psychopharmacology, Central Institute of Mental Health, Faculty of Medicine, University of Heidelberg, Mannheim, Germany
| | - Marcela Hernández
- Laboratory of Periodontal Biology, Faculty of Dentistry, Universidad de Chile, Santiago, Chile.,Department of Pathology and Oral Medicine, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| |
Collapse
|
8
|
Bhuiyan MIH, Young CB, Jahan I, Hasan MN, Fischer S, Meor Azlan NF, Liu M, Chattopadhyay A, Huang H, Kahle KT, Zhang J, Poloyac SM, Molyneaux BJ, Straub AC, Deng X, Gomez D, Sun D. NF-κB Signaling-Mediated Activation of WNK-SPAK-NKCC1 Cascade in Worsened Stroke Outcomes of Ang II-Hypertensive Mice. Stroke 2022; 53:1720-1734. [PMID: 35272484 PMCID: PMC9038703 DOI: 10.1161/strokeaha.121.038351] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 01/31/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND Worsened stroke outcomes with hypertension comorbidity are insensitive to blood pressure-lowering therapies. In an experimental stroke model with comorbid hypertension, we investigated causal roles of ang II (angiotensin II)-mediated stimulation of the brain WNK (with no lysine [K] kinases)-SPAK (STE20/SPS1-related proline/alanine-rich kinase)-NKCC1 (Na-K-Cl cotransporter) complex in worsened outcomes. METHODS Saline- or ang II-infused C57BL/6J male mice underwent stroke induced by permanent occlusion of the distal branches of the middle cerebral artery. Mice were randomly assigned to receive either vehicle dimethyl sulfoxide/PBS (2 mL/kg body weight/day, IP), a novel SPAK inhibitor, 5-chloro-N-(5-chloro-4-((4-chlorophenyl)(cyano)methyl)-2-methylphenyl)-2-hydroxybenzamide (ZT-1a' 5 mg/kg per day, IP) or a NF-κB (nuclear factor-κB) inhibitor TAT-NBD (transactivator of transcription-NEMO-binding domain' 20 mg/kg per day, IP). Activation of brain NF-κB and WNK-SPAK-NKCC1 cascade as well as ischemic stroke outcomes were examined. RESULTS Stroke triggered a 2- to 5-fold increase of WNK (isoforms 1, 2, 4), SPAK/OSR1 (oxidative stress-responsive kinase 1), and NKCC1 protein in the ang II-infused hypertensive mouse brains at 24 hours after stroke, which was associated with increased nuclear translocation of phospho-NF-κB protein in the cortical neurons (a Pearson correlation r of 0.77, P<0.005). The upregulation of WNK-SPAK-NKCC1 cascade proteins resulted from increased NF-κB recruitment on Wnk1, Wnk2, Wnk4, Spak, and Nkcc1 gene promoters and was attenuated by NF-κB inhibitor TAT-NBD. Poststroke administration of SPAK inhibitor ZT-1a significantly reduced WNK-SPAK-NKCC1 complex activation, brain lesion size, and neurological function deficits in the ang II-hypertensive mice without affecting blood pressure and cerebral blood flow. CONCLUSIONS The ang II-induced stimulation of NF-κB transcriptional activity upregulates brain WNK-SPAK-NKCC1 cascade and contributes to worsened ischemic stroke outcomes, illustrating the brain WNK-SPAK-NKCC1 complex as a therapeutic target for stroke with comorbid hypertension.
Collapse
Affiliation(s)
- Mohammad Iqbal H Bhuiyan
- Departments of Neurology (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., H.H., B.J.M., D.S.), University of Pittsburgh, PA
- Pittsburgh Institute for Neurodegenerative Disorders (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., B.J.M., D.S.), University of Pittsburgh, PA
- Veterans Affairs Pittsburgh Health Care System, Geriatric Research, Educational, and Clinical Center, PA (M.I.H.B.' D.S.)
| | - Cullen B Young
- Departments of Neurology (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., H.H., B.J.M., D.S.), University of Pittsburgh, PA
- Pittsburgh Institute for Neurodegenerative Disorders (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., B.J.M., D.S.), University of Pittsburgh, PA
| | - Israt Jahan
- Departments of Neurology (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., H.H., B.J.M., D.S.), University of Pittsburgh, PA
- Pittsburgh Institute for Neurodegenerative Disorders (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., B.J.M., D.S.), University of Pittsburgh, PA
| | - Md Nabiul Hasan
- Departments of Neurology (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., H.H., B.J.M., D.S.), University of Pittsburgh, PA
- Pittsburgh Institute for Neurodegenerative Disorders (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., B.J.M., D.S.), University of Pittsburgh, PA
| | - Sydney Fischer
- Departments of Neurology (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., H.H., B.J.M., D.S.), University of Pittsburgh, PA
- Pittsburgh Institute for Neurodegenerative Disorders (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., B.J.M., D.S.), University of Pittsburgh, PA
| | - Nur Farah Meor Azlan
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, United Kingdom (N.F.M.A., J.Z.)
| | - Mingjun Liu
- Medicine (M.L., D.G.), University of Pittsburgh, PA
| | - Ansuman Chattopadhyay
- Molecular Biology-Information Service, Health Sciences Library System (A.C.), University of Pittsburgh, PA
| | - Huachen Huang
- Departments of Neurology (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., H.H., B.J.M., D.S.), University of Pittsburgh, PA
| | - Kristopher T Kahle
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston (K.T.K.)
| | - Jinwei Zhang
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, United Kingdom (N.F.M.A., J.Z.)
| | | | - Bradley J Molyneaux
- Departments of Neurology (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., H.H., B.J.M., D.S.), University of Pittsburgh, PA
- Pittsburgh Institute for Neurodegenerative Disorders (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., B.J.M., D.S.), University of Pittsburgh, PA
| | - Adam C Straub
- Pharmacology and Chemical Biology (A.C.S), University of Pittsburgh, PA
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute (A.C.S., D.G.), University of Pittsburgh, PA
| | - Xianming Deng
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Fujian, China (X.D.)
| | - Delphine Gomez
- Medicine (M.L., D.G.), University of Pittsburgh, PA
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute (A.C.S., D.G.), University of Pittsburgh, PA
| | - Dandan Sun
- Departments of Neurology (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., H.H., B.J.M., D.S.), University of Pittsburgh, PA
- Pittsburgh Institute for Neurodegenerative Disorders (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., B.J.M., D.S.), University of Pittsburgh, PA
- Veterans Affairs Pittsburgh Health Care System, Geriatric Research, Educational, and Clinical Center, PA (M.I.H.B.' D.S.)
| |
Collapse
|
9
|
Wang J, Liu R, Hasan MN, Fischer S, Chen Y, Como M, Fiesler VM, Bhuiyan MIH, Dong S, Li E, Kahle KT, Zhang J, Deng X, Subramanya AR, Begum G, Yin Y, Sun D. Role of SPAK-NKCC1 signaling cascade in the choroid plexus blood-CSF barrier damage after stroke. J Neuroinflammation 2022; 19:91. [PMID: 35413993 PMCID: PMC9006540 DOI: 10.1186/s12974-022-02456-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/29/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The mechanisms underlying dysfunction of choroid plexus (ChP) blood-cerebrospinal fluid (CSF) barrier and lymphocyte invasion in neuroinflammatory responses to stroke are not well understood. In this study, we investigated whether stroke damaged the blood-CSF barrier integrity due to dysregulation of major ChP ion transport system, Na+-K+-Cl- cotransporter 1 (NKCC1), and regulatory Ste20-related proline-alanine-rich kinase (SPAK). METHODS Sham or ischemic stroke was induced in C57Bl/6J mice. Changes on the SPAK-NKCC1 complex and tight junction proteins (TJs) in the ChP were quantified by immunofluorescence staining and immunoblotting. Immune cell infiltration in the ChP was assessed by flow cytometry and immunostaining. Cultured ChP epithelium cells (CPECs) and cortical neurons were used to evaluate H2O2-mediated oxidative stress in stimulating the SPAK-NKCC1 complex and cellular damage. In vivo or in vitro pharmacological blockade of the ChP SPAK-NKCC1 cascade with SPAK inhibitor ZT-1a or NKCC1 inhibitor bumetanide were examined. RESULTS Ischemic stroke stimulated activation of the CPECs apical membrane SPAK-NKCC1 complex, NF-κB, and MMP9, which was associated with loss of the blood-CSF barrier integrity and increased immune cell infiltration into the ChP. Oxidative stress directly activated the SPAK-NKCC1 pathway and resulted in apoptosis, neurodegeneration, and NKCC1-mediated ion influx. Pharmacological blockade of the SPAK-NKCC1 pathway protected the ChP barrier integrity, attenuated ChP immune cell infiltration or neuronal death. CONCLUSION Stroke-induced pathological stimulation of the SPAK-NKCC1 cascade caused CPECs damage and disruption of TJs at the blood-CSF barrier. The ChP SPAK-NKCC1 complex emerged as a therapeutic target for attenuating ChP dysfunction and lymphocyte invasion after stroke.
Collapse
Affiliation(s)
- Jun Wang
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian, 116027, Liaoning, China
- Department of Neurology, University of Pittsburgh, 7016 Biomedical Science Tower 3, 3501 Fifth Ave., Pittsburgh, PA, 15260, USA
| | - Ruijia Liu
- Department of Neurology, University of Pittsburgh, 7016 Biomedical Science Tower 3, 3501 Fifth Ave., Pittsburgh, PA, 15260, USA
| | - Md Nabiul Hasan
- Department of Neurology, University of Pittsburgh, 7016 Biomedical Science Tower 3, 3501 Fifth Ave., Pittsburgh, PA, 15260, USA
- Research Service, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA
| | - Sydney Fischer
- Department of Neurology, University of Pittsburgh, 7016 Biomedical Science Tower 3, 3501 Fifth Ave., Pittsburgh, PA, 15260, USA
| | - Yang Chen
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian, 116027, Liaoning, China
- Department of Neurology, University of Pittsburgh, 7016 Biomedical Science Tower 3, 3501 Fifth Ave., Pittsburgh, PA, 15260, USA
| | - Matt Como
- Pennsylvania State University, State College, PA, USA
| | - Victoria M Fiesler
- Department of Neurology, University of Pittsburgh, 7016 Biomedical Science Tower 3, 3501 Fifth Ave., Pittsburgh, PA, 15260, USA
| | - Mohammad Iqbal H Bhuiyan
- Department of Neurology, University of Pittsburgh, 7016 Biomedical Science Tower 3, 3501 Fifth Ave., Pittsburgh, PA, 15260, USA
- Research Service, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA
| | - Shuying Dong
- Department of Neurology, University of Pittsburgh, 7016 Biomedical Science Tower 3, 3501 Fifth Ave., Pittsburgh, PA, 15260, USA
| | - Eric Li
- Department of Neurology, University of Pittsburgh, 7016 Biomedical Science Tower 3, 3501 Fifth Ave., Pittsburgh, PA, 15260, USA
| | - Kristopher T Kahle
- Department of Neurosurgery, The Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jinwei Zhang
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, Hatherly Laboratory, Exeter, EX4 4PS, UK
| | - Xianming Deng
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Arohan R Subramanya
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Research Service, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA
| | - Gulnaz Begum
- Department of Neurology, University of Pittsburgh, 7016 Biomedical Science Tower 3, 3501 Fifth Ave., Pittsburgh, PA, 15260, USA
- Research Service, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA
| | - Yan Yin
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian, 116027, Liaoning, China.
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh, 7016 Biomedical Science Tower 3, 3501 Fifth Ave., Pittsburgh, PA, 15260, USA.
- Research Service, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA.
| |
Collapse
|
10
|
Huang TY, Yang SS, Liao CL, Lin MH, Lin HH, Lin JC, Chen PJ, Shih YL, Chang WK, Hsieh TY. SPAK Deficiency Attenuates Chemotherapy-Induced Intestinal Mucositis. Front Oncol 2021; 11:733555. [PMID: 34888232 PMCID: PMC8649624 DOI: 10.3389/fonc.2021.733555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 11/03/2021] [Indexed: 11/13/2022] Open
Abstract
Introduction Ste20-related protein proline/alanine-rich kinase (SPAK) affects cell proliferation, differentiation, and transformation, and sodium and chloride transport in the gut. However, its role in gut injury pathogenesis is unclear. Objective We determined the role of SPAK in chemotherapy-induced intestinal mucositis using in vivo and in vitro models. Methods Using SPAK-knockout (KO) mice, we evaluated the severity of intestinal mucositis induced by 5-fluorouracil (5-FU) by assessing body weight loss, histological changes in the intestinal mucosa, length of villi in the small intestine, pro-inflammatory cytokine levels, proliferative indices, and apoptotic indices. We also evaluated changes in gut permeability and tight junction-associated protein expression. Changes in cell permeability, proliferation, and apoptosis were assessed in SPAK siRNA-transfected 5FU-treated IEC-6 cells. Results 5-FU-treated SPAK-KO mice exhibited milder intestinal mucositis, reduced pro-inflammatory cytokine expression, increased villus length, good maintenance of proliferative indices of villus cells, decreased apoptotic index of enterocytes, reduced gut permeability, and restoration of tight junction protein expression (vs. 5-FU-treated wild-type mice). Under in vitro conditions, siRNA-mediated SPAK-knockdown in IEC-6 cells decreased cell permeability and maintained homeostasis following 5-FU treatment. Conclusion SPAK deficiency attenuated chemotherapy-induced intestinal mucositis by modulating gut permeability and tight junction-associated protein expression and maintaining gut homeostasis in murine small intestinal tissues following gut injury. The expression of SPAK may influence the pathogenesis of chemotherapy-induced intestinal mucositis.
Collapse
Affiliation(s)
- Tien-Yu Huang
- Division of Gastroenterology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Taiwan Association for the Study of Small Intestinal Diseases, Taoyuan, Taiwan
| | - Sung-Sen Yang
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.,Institute of BioMedical Science, Academia Sinica, Taipei, Taiwan
| | - Ching-Len Liao
- Department of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Ming-Hong Lin
- Department of Microbiology and Immunology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hsuan-Hwai Lin
- Division of Gastroenterology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Jung-Chun Lin
- Division of Gastroenterology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Peng-Jen Chen
- Division of Gastroenterology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Lueng Shih
- Division of Gastroenterology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Wei-Kuo Chang
- Division of Gastroenterology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Tsai-Yuan Hsieh
- Division of Gastroenterology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
11
|
Cui J, Xu H, Lehtinen MK. Macrophages on the margin: choroid plexus immune responses. Trends Neurosci 2021; 44:864-875. [PMID: 34312005 PMCID: PMC8551004 DOI: 10.1016/j.tins.2021.07.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 12/11/2022]
Abstract
The choroid plexus (ChP), an epithelial bilayer containing a network of mesenchymal, immune, and neuronal cells, forms the blood-cerebrospinal fluid (CSF) barrier (BCSFB). While best recognized for secreting CSF, the ChP is also a hotbed of immune cell activity and can provide circulating peripheral immune cells with passage into the central nervous system (CNS). Here, we review recent studies on ChP immune cells, with a focus on the ontogeny, development, and behaviors of ChP macrophages, the principal resident immune cells of the ChP. We highlight the implications of immune cells for ChP barrier function, CSF cytokines and volume regulation, and their contribution to neurodevelopmental disorders, with possible age-specific features to be elucidated in the future.
Collapse
Affiliation(s)
- Jin Cui
- Department of Pathology, Boston Children's Hospital, Boston, MA, USA
| | - Huixin Xu
- Department of Pathology, Boston Children's Hospital, Boston, MA, USA
| | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
12
|
Karimy JK, Reeves BC, Damisah E, Duy PQ, Antwi P, David W, Wang K, Schiff SJ, Limbrick DD, Alper SL, Warf BC, Nedergaard M, Simard JM, Kahle KT. Inflammation in acquired hydrocephalus: pathogenic mechanisms and therapeutic targets. Nat Rev Neurol 2020; 16:285-296. [PMID: 32152460 DOI: 10.1038/s41582-020-0321-y] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2020] [Indexed: 12/11/2022]
Abstract
Hydrocephalus is the most common neurosurgical disorder worldwide and is characterized by enlargement of the cerebrospinal fluid (CSF)-filled brain ventricles resulting from failed CSF homeostasis. Since the 1840s, physicians have observed inflammation in the brain and the CSF spaces in both posthaemorrhagic hydrocephalus (PHH) and postinfectious hydrocephalus (PIH). Reparative inflammation is an important protective response that eliminates foreign organisms, damaged cells and physical irritants; however, inappropriately triggered or sustained inflammation can respectively initiate or propagate disease. Recent data have begun to uncover the molecular mechanisms by which inflammation - driven by Toll-like receptor 4-regulated cytokines, immune cells and signalling pathways - contributes to the pathogenesis of hydrocephalus. We propose that therapeutic approaches that target inflammatory mediators in both PHH and PIH could address the multiple drivers of disease, including choroid plexus CSF hypersecretion, ependymal denudation, and damage and scarring of intraventricular and parenchymal (glia-lymphatic) CSF pathways. Here, we review the evidence for a prominent role of inflammation in the pathogenic mechanism of PHH and PIH and highlight promising targets for therapeutic intervention. Focusing research efforts on inflammation could shift our view of hydrocephalus from that of a lifelong neurosurgical disorder to that of a preventable neuroinflammatory condition.
Collapse
Affiliation(s)
- Jason K Karimy
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Benjamin C Reeves
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Eyiyemisi Damisah
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Phan Q Duy
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Prince Antwi
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Wyatt David
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Kevin Wang
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Steven J Schiff
- Departments of Neurosurgery, Engineering Science & Mechanics, and Physics; Center for Neural Engineering, The Pennsylvania State University, University Park, PA, USA
| | - David D Limbrick
- Departments of Neurosurgery and Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Seth L Alper
- Division of Nephrology and Vascular Biology Research Center, Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Benjamin C Warf
- Department of Neurosurgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA.,Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - J Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kristopher T Kahle
- Departments of Neurosurgery, Pediatrics, and Cellular & Molecular Physiology and Yale-Rockefeller NIH Centers for Mendelian Genomics, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
13
|
Hung CM, Peng CK, Yang SS, Shui HA, Huang KL. WNK4–SPAK modulates lipopolysaccharide-induced macrophage activation. Biochem Pharmacol 2020; 171:113738. [DOI: 10.1016/j.bcp.2019.113738] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 11/26/2019] [Indexed: 01/23/2023]
|
14
|
Gallolu Kankanamalage S, Karra AS, Cobb MH. WNK pathways in cancer signaling networks. Cell Commun Signal 2018; 16:72. [PMID: 30390653 PMCID: PMC6215617 DOI: 10.1186/s12964-018-0287-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 10/22/2018] [Indexed: 12/13/2022] Open
Abstract
Background The with no lysine [K] (WNK) pathway consists of the structurally unique WNK kinases, their downstream target kinases, oxidative stress responsive (OSR)1 and SPS/Ste20-related proline-alanine-rich kinase (SPAK), and a multitude of OSR1/SPAK substrates including cation chloride cotransporters. Main body While the best known functions of the WNK pathway is regulation of ion transport across cell membranes, WNK pathway components have been implicated in numerous human diseases. The goal of our review is to draw attention to how this pathway and its components exert influence on the progression of cancer, specifically by detailing WNK signaling intersections with major cell communication networks and processes. Conclusion Here we describe how WNKs and associated proteins interact with and influence PI3K-AKT, TGF-β, and NF-κB signaling, as well as its unanticipated role in the regulation of angiogenesis.
Collapse
Affiliation(s)
- Sachith Gallolu Kankanamalage
- Department of Pharmacology, The University of Texas Southwestern Medical Center, 6001 Forest Park Road, Dallas, TX, 75390-9041, USA
| | - Aroon S Karra
- Department of Pharmacology, The University of Texas Southwestern Medical Center, 6001 Forest Park Road, Dallas, TX, 75390-9041, USA
| | - Melanie H Cobb
- Department of Pharmacology, The University of Texas Southwestern Medical Center, 6001 Forest Park Road, Dallas, TX, 75390-9041, USA.
| |
Collapse
|
15
|
Yang H, Mao W, Rodriguez-Aguayo C, Mangala LS, Bartholomeusz G, Iles LR, Jennings NB, Ahmed AA, Sood AK, Lopez-Berestein G, Lu Z, Bast RC. Paclitaxel Sensitivity of Ovarian Cancer Can be Enhanced by Knocking Down Pairs of Kinases that Regulate MAP4 Phosphorylation and Microtubule Stability. Clin Cancer Res 2018; 24:5072-5084. [PMID: 30084832 DOI: 10.1158/1078-0432.ccr-18-0504] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 05/08/2018] [Accepted: 06/27/2018] [Indexed: 01/11/2023]
Abstract
Purpose: Most patients with ovarian cancer receive paclitaxel chemotherapy, but less than half respond. Pre-treatment microtubule stability correlates with paclitaxel response in ovarian cancer cell lines. Microtubule stability can be increased by depletion of individual kinases. As microtubule stability can be regulated by phosphorylation of microtubule-associated proteins (MAPs), we reasoned that depletion of pairs of kinases that regulate phosphorylation of MAPs could induce microtubule stabilization and paclitaxel sensitization.Experimental Design: Fourteen kinases known to regulate paclitaxel sensitivity were depleted individually in 12 well-characterized ovarian cancer cell lines before measuring proliferation in the presence or absence of paclitaxel. Similar studies were performed by depleting all possible pairs of kinases in six ovarian cancer cell lines. Pairs that enhanced paclitaxel sensitivity across multiple cell lines were studied in depth in cell culture and in two xenograft models.Results: Transfection of siRNA against 10 of the 14 kinases enhanced paclitaxel sensitivity in at least six of 12 cell lines. Dual knockdown of IKBKB/STK39 or EDN2/TBK1 enhanced paclitaxel sensitivity more than silencing single kinases. Sequential knockdown was superior to concurrent knockdown. Dual silencing of IKBKB/STK39 or EDN2/TBK1 stabilized microtubules by inhibiting phosphorylation of p38 and MAP4, inducing apoptosis and blocking cell cycle more effectively than silencing individual kinases. Knockdown of IKBKB/STK39 or EDN2/TBK1 enhanced paclitaxel sensitivity in two ovarian xenograft models.Conclusions: Sequential knockdown of dual kinases increased microtubule stability by decreasing p38-mediated phosphorylation of MAP4 and enhanced response to paclitaxel in ovarian cancer cell lines and xenografts, suggesting a strategy to improve primary therapy. Clin Cancer Res; 24(20); 5072-84. ©2018 AACR.
Collapse
Affiliation(s)
- Hailing Yang
- Department of Experimental Therapeutics, University of Texas, M.D. Anderson Cancer Center, Houston, Texas
| | - Weiqun Mao
- Department of Experimental Therapeutics, University of Texas, M.D. Anderson Cancer Center, Houston, Texas
| | - Cristian Rodriguez-Aguayo
- Department of Experimental Therapeutics, University of Texas, M.D. Anderson Cancer Center, Houston, Texas.,Center for RNA Interference and Non-Coding RNA, University of Texas, M.D. Anderson Cancer Center, Houston, Texas
| | - Lingegowda S Mangala
- Center for RNA Interference and Non-Coding RNA, University of Texas, M.D. Anderson Cancer Center, Houston, Texas.,Department of Gynecologic Oncology and Reproductive Medicine, University of Texas, M.D. Anderson Cancer Center, Houston, Texas
| | - Geoffrey Bartholomeusz
- Department of Experimental Therapeutics, University of Texas, M.D. Anderson Cancer Center, Houston, Texas
| | - Lakesla R Iles
- Department of Experimental Therapeutics, University of Texas, M.D. Anderson Cancer Center, Houston, Texas
| | - Nicholas B Jennings
- Center for RNA Interference and Non-Coding RNA, University of Texas, M.D. Anderson Cancer Center, Houston, Texas
| | - Ahmed Ashour Ahmed
- Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, United Kingdom.,Nuffield Department of Women's & Reproductive Health, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, United Kingdom
| | - Anil K Sood
- Center for RNA Interference and Non-Coding RNA, University of Texas, M.D. Anderson Cancer Center, Houston, Texas.,Department of Gynecologic Oncology and Reproductive Medicine, University of Texas, M.D. Anderson Cancer Center, Houston, Texas
| | - Gabriel Lopez-Berestein
- Department of Experimental Therapeutics, University of Texas, M.D. Anderson Cancer Center, Houston, Texas.,Center for RNA Interference and Non-Coding RNA, University of Texas, M.D. Anderson Cancer Center, Houston, Texas
| | - Zhen Lu
- Department of Experimental Therapeutics, University of Texas, M.D. Anderson Cancer Center, Houston, Texas
| | - Robert C Bast
- Department of Experimental Therapeutics, University of Texas, M.D. Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
16
|
Effects of SPAK on vascular reactivity and nitric oxide production in endotoxemic mice. Eur J Pharmacol 2017; 814:248-254. [PMID: 28864211 DOI: 10.1016/j.ejphar.2017.08.038] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 08/21/2017] [Accepted: 08/28/2017] [Indexed: 11/20/2022]
Abstract
Vasoplegia impedes therapeutic interventions to restore vascular tone, leading to severe hypotension, poor tissue perfusion, and multiple organ failure in septic shock. High levels of circulating nitric oxide (NO) play a crucial role in endotoxin-induced vascular hyporeactivity. Proinflammatory cytokines have been implicated in the induction of inducible NO synthase and overproduction of NO. Anti-inflammatory therapy can diminish NO formation and improve vascular hyporeactivity in septic shock. STE20/SPS1-realted proline/alanine-rich kinase (SPAK) has been reported to activate mitogen-activated protein kinase and contribute to intestinal inflammation. Thus, we evaluated the roles of SPAK in NO production and vascular hyporeactivity in endotoxemic animals. Male wild-type and SPAK deficiency mice were intraperitoneally administered vehicle or Escherichia coli lipopolysaccharide (LPS, 50mg/kg). The changes of systolic blood pressure and plasma nitrate and nitrite levels were measured during the experimental period. Thoracic aortas were exercised to assess vascular reactivity and SPAK expression. In the present study, mice in endotoxin model showed severe hypotension and hyporeactivity to serotonin, phenylephrine (PE), and acetylcholine in the aortic rings. Phosphorylated SPAK expression in the aorta and NO levels in the plasma were also increased in animals with endotoxic shock. However, deletion of SPAK not only reduced the elevation of NO levels but also improved vascular hyporeactivity to serotonin and PE in endotoxemic mice. Taken together, SPAK could be involved in the NO overproduction and vascular hyporesponsiveness to vasoconstrictors in endotoxic shock. Thus, inhibition of SPAK could be useful in the prevention of endotoxin-induced vascular hyporeactivity.
Collapse
|
17
|
Karimy JK, Zhang J, Kurland DB, Theriault BC, Duran D, Stokum JA, Furey CG, Zhou X, Mansuri MS, Montejo J, Vera A, DiLuna ML, Delpire E, Alper SL, Gunel M, Gerzanich V, Medzhitov R, Simard JM, Kahle KT. Inflammation-dependent cerebrospinal fluid hypersecretion by the choroid plexus epithelium in posthemorrhagic hydrocephalus. Nat Med 2017; 23:997-1003. [PMID: 28692063 DOI: 10.1038/nm.4361] [Citation(s) in RCA: 256] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Accepted: 05/31/2017] [Indexed: 02/07/2023]
Abstract
The choroid plexus epithelium (CPE) secretes higher volumes of fluid (cerebrospinal fluid, CSF) than any other epithelium and simultaneously functions as the blood-CSF barrier to gate immune cell entry into the central nervous system. Posthemorrhagic hydrocephalus (PHH), an expansion of the cerebral ventricles due to CSF accumulation following intraventricular hemorrhage (IVH), is a common disease usually treated by suboptimal CSF shunting techniques. PHH is classically attributed to primary impairments in CSF reabsorption, but little experimental evidence supports this concept. In contrast, the potential contribution of CSF secretion to PHH has received little attention. In a rat model of PHH, we demonstrate that IVH causes a Toll-like receptor 4 (TLR4)- and NF-κB-dependent inflammatory response in the CPE that is associated with a ∼3-fold increase in bumetanide-sensitive CSF secretion. IVH-induced hypersecretion of CSF is mediated by TLR4-dependent activation of the Ste20-type stress kinase SPAK, which binds, phosphorylates, and stimulates the NKCC1 co-transporter at the CPE apical membrane. Genetic depletion of TLR4 or SPAK normalizes hyperactive CSF secretion rates and reduces PHH symptoms, as does treatment with drugs that antagonize TLR4-NF-κB signaling or the SPAK-NKCC1 co-transporter complex. These data uncover a previously unrecognized contribution of CSF hypersecretion to the pathogenesis of PHH, demonstrate a new role for TLRs in regulation of the internal brain milieu, and identify a kinase-regulated mechanism of CSF secretion that could be targeted by repurposed US Food and Drug Administration (FDA)-approved drugs to treat hydrocephalus.
Collapse
Affiliation(s)
- Jason K Karimy
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Jinwei Zhang
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratory, Exeter, UK
| | - David B Kurland
- Department of Neurosurgery, University of Maryland, School of Medicine, Baltimore, Maryland, USA
| | | | - Daniel Duran
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Jesse A Stokum
- Department of Neurosurgery, University of Maryland, School of Medicine, Baltimore, Maryland, USA
| | | | - Xu Zhou
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - M Shahid Mansuri
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Julio Montejo
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Alberto Vera
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Michael L DiLuna
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Seth L Alper
- Division of Nephrology, Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Vascular Biology Research Center, Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Murat Gunel
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland, School of Medicine, Baltimore, Maryland, USA
| | - Ruslan Medzhitov
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - J Marc Simard
- Department of Neurosurgery, University of Maryland, School of Medicine, Baltimore, Maryland, USA
- Department of Pathology, University of Maryland, School of Medicine, Baltimore, Maryland, USA
- Department of Physiology, University of Maryland, School of Medicine, Baltimore, Maryland, USA
| | - Kristopher T Kahle
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, USA
- Center for Mendelian Genomics, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
18
|
Zhang J, Karimy JK, Delpire E, Kahle KT. Pharmacological targeting of SPAK kinase in disorders of impaired epithelial transport. Expert Opin Ther Targets 2017; 21:795-804. [PMID: 28679296 PMCID: PMC6081737 DOI: 10.1080/14728222.2017.1351949] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The mammalian SPS1-related proline/alanine-rich serine-threonine kinase SPAK (STK39) modulates ion transport across and between epithelial cells in response to environmental stimuli such osmotic stress and inflammation. Research over the last decade has established a central role for SPAK in the regulation of ion and water transport in the distal nephron, colonic crypts, and pancreatic ducts, and has implicated deregulated SPAK signaling in NaCl-sensitive hypertension, ulcerative colitis and Crohn's disease, and cystic fibrosis. Areas covered: We review recent advances in our understanding of the role of SPAK kinase in the regulation of epithelial transport. We highlight how SPAK signaling - including its upstream Cl- sensitive activators, the WNK kinases, and its downstream ion transport targets, the cation- Cl- cotransporters contribute to human disease. We discuss prospects for the pharmacotherapeutic targeting of SPAK kinase in specific human disorders that feature impaired epithelial homeostasis. Expert opinion: The development of novel drugs that antagonize the SPAK-WNK interaction, inhibit SPAK kinase activity, or disrupt SPAK kinase activation by interfering with its binding to MO25α/β could be useful adjuncts in essential hypertension, inflammatory colitis, and cystic fibrosis.
Collapse
Affiliation(s)
- Jinwei Zhang
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratory, Exeter, EX4 4PS, UK
| | - Jason K. Karimy
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06510, USA
| | - Eric Delpire
- Department of Anesthesiolgy, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Kristopher T. Kahle
- Departments of Neurosurgery, Pediatrics, and Cellular & Molecular Physiology; and Centers for Mendelian Genomics, Yale School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
19
|
Abstract
WNK kinases, along with their upstream regulators (CUL3/KLHL3) and downstream targets (the SPAK/OSR1 kinases and the cation-Cl- cotransporters [CCCs]), comprise a signaling cascade essential for ion homeostasis in the kidney and nervous system. Recent work has furthered our understanding of the WNKs in epithelial transport, cell volume homeostasis, and GABA signaling, and uncovered novel roles for this pathway in immune cell function and cell proliferation.
Collapse
Affiliation(s)
- Masoud Shekarabi
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Jinwei Zhang
- Departments of Neurosurgery, Centers for Mendelian Genomics, Yale School of Medicine, New Haven, CT 06477, USA; MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, UK
| | - Arjun R Khanna
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Neurosurgery, Harvard Medical School, Boston, MA 02115, USA
| | - David H Ellison
- Division of Nephrology & Hypertension, Department of Medicine, Oregon Health & Science University, Portland, Oregon 97239, USA; VA Portland Health Care System, Portland, OR 97239, USA
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Kristopher T Kahle
- Departments of Neurosurgery, Pediatrics, and Cellular & Molecular Physiology, Centers for Mendelian Genomics, Yale School of Medicine, New Haven, CT 06477, USA.
| |
Collapse
|
20
|
Lin TJ, Yang SS, Hua KF, Tsai YL, Lin SH, Ka SM. SPAK plays a pathogenic role in IgA nephropathy through the activation of NF-κB/MAPKs signaling pathway. Free Radic Biol Med 2016; 99:214-224. [PMID: 27519267 DOI: 10.1016/j.freeradbiomed.2016.08.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Revised: 08/05/2016] [Accepted: 08/08/2016] [Indexed: 01/13/2023]
Abstract
Sterile 20/SPS1-related proline/alanine-rich kinase (SPAK) can stimulate production of proinflammatory cytokines and interact with inflammation-related molecules. However, it has yet to be determined whether SPAK plays a pathophysiological role in the complicated pathological mechanisms of IgA nephropathy (IgAN), which is mainly characterized by mesangial cell (MC) proliferation and is the most common form of glomerulonephritis. In the present study, we examined the pathophysiological role of SPAK in IgAN using a mouse model and cell models. Our results clearly showed that (1) SPAK deficiency prevents the development of IgAN and inhibits production of immune/inflammatory mediators and T cell activation and proliferation; and (2) when primed with IgA immune complexes (IgA IC), both peritoneal macrophages and primary MCs from SPAK knockout mice show markedly reduced production of proinflammatory cytokines and inhibition of NF-κB/MAPKs activation. We proposed that activation of SPAK and the NF-κB/MAPKs signaling pathway in MCs, macrophages and T cells of the glomerulus may be a mechanism underlying the pathogenesis of IgAN. The activation of SPAK in renal tubuloepithelial cells either directly by IgA IC or an indirect action of the activated MCs or infiltrating mononuclear leukocytes seen in the kidney may further aggravate the disease process of IgAN. Our results suggest that SPAK is a potential therapeutic target for the glomerular disorder.
Collapse
Affiliation(s)
- Tsai-Jung Lin
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Sung-Sen Yang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan; Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan; Institute of BioMedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Kuo-Feng Hua
- Department of Biotechnology and Animal Science, National Ilan University, Ilan, Taiwan
| | - Yu-Ling Tsai
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Hua Lin
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shuk-Man Ka
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan; Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan; Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
21
|
With-No-Lysine Kinase 4 Mediates Alveolar Fluid Regulation in Hyperoxia-Induced Lung Injury. Crit Care Med 2015; 43:e412-9. [PMID: 26035408 DOI: 10.1097/ccm.0000000000001144] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVES To investigate mechanisms involved in the regulation of epithelial ion channels and alveolar fluid clearance in hyperoxia-induced lung injury. DESIGN Laboratory animal experiments. SETTING Animal care facility procedure room in a medical center. SUBJECTS Wild-type, STE20/SPS1-related proline/alanine-rich kinase knockout (SPAK(-/-)), and with-no-lysine kinase 4 knockin (WNK4(D561A/+)) mice. INTERVENTIONS Mice were exposed to room air or 95% hyperoxia for 60 hours. MEASUREMENTS AND MAIN RESULTS Exposure to hyperoxia for 60 hours increased the lung expression of with-no-lysine kinase 4 and led to STE20/SPS1-related proline/alanine-rich kinase and sodium-potassium-chloride cotransporter phosphorylation, which resulted in the suppression of alveolar fluid clearance and increase of lung edema. WNK4(D561A/+) mice at the baseline presented an abundance of epithelium sodium channel and high levels of STE20/SPS1-related proline/alanine-rich kinase and sodium-potassium-chloride cotransporter phosphorylation. Compared with the wild-type group, hyperoxia caused greater epithelium sodium channel expression in WNK4(D561A/+) mice, but no significant difference in STE20/SPS1-related proline/alanine-rich kinase and sodium-potassium-chloride cotransporter phosphorylation. The functional inactivation of sodium-potassium-chloride cotransporter by gene knockout in SPAK(-/-) mice yielded a lower severity of lung injury and longer animal survival, whereas constitutive expression of with-no-lysine kinase 4 exacerbated the hyperoxia-induced lung injury. Pharmacologic inhibition of sodium-potassium-chloride cotransporter by inhaled furosemide improved animal survival in WNK4(D561A/+) mice. By contrast, inhibition of epithelium sodium channel exacerbated the hyperoxia-induced lung injury and animal death. CONCLUSIONS With-no-lysine kinase 4 plays a crucial role in the regulation of epithelial ion channels and alveolar fluid clearance, mainly via phosphorylation and activation of STE20/SPS1-related proline/alanine-rich kinase and sodium-potassium-chloride cotransporter.
Collapse
|
22
|
Huang Y, Hu N, Gao X, Yan Z, Li S, Jing W, Yan R. Alterations of testosterone metabolism in microsomes from rats with experimental colitis induced by dextran sulfate sodium. Chem Biol Interact 2015; 232:38-48. [DOI: 10.1016/j.cbi.2015.02.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 02/13/2015] [Accepted: 02/17/2015] [Indexed: 01/22/2023]
|
23
|
Centanni M, Turroni S, Rampelli S, Biagi E, Quercia S, Consolandi C, Severgnini M, Brigidi P, Candela M. Bifidobacterium animalisssp.lactisBI07 modulates the tumor necrosis factor alpha-dependent imbalances of the enterocyte-associated intestinal microbiota fraction. FEMS Microbiol Lett 2014; 357:157-63. [DOI: 10.1111/1574-6968.12515] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 06/06/2014] [Accepted: 06/18/2014] [Indexed: 12/12/2022] Open
Affiliation(s)
- Manuela Centanni
- Department of Pharmacy and Biotechnology; University of Bologna; Bologna Italy
| | - Silvia Turroni
- Department of Pharmacy and Biotechnology; University of Bologna; Bologna Italy
| | - Simone Rampelli
- Department of Pharmacy and Biotechnology; University of Bologna; Bologna Italy
| | - Elena Biagi
- Department of Pharmacy and Biotechnology; University of Bologna; Bologna Italy
| | - Sara Quercia
- Department of Pharmacy and Biotechnology; University of Bologna; Bologna Italy
| | - Clarissa Consolandi
- Institute of Biomedical Technologies - Italian National Research Council; Milan Italy
| | - Marco Severgnini
- Institute of Biomedical Technologies - Italian National Research Council; Milan Italy
| | - Patrizia Brigidi
- Department of Pharmacy and Biotechnology; University of Bologna; Bologna Italy
| | - Marco Candela
- Department of Pharmacy and Biotechnology; University of Bologna; Bologna Italy
| |
Collapse
|
24
|
Tan X, Li D, Wang X, Zeng Y, Yan Y, Yang L. Claudin-2 downregulation by KSHV infection is involved in the regulation of endothelial barrier function. J Cutan Pathol 2014; 41:630-9. [PMID: 24995964 DOI: 10.1111/cup.12332] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 02/01/2014] [Accepted: 02/17/2014] [Indexed: 12/25/2022]
Abstract
BACKGROUND Kaposi sarcoma (KS), caused by the infection of Kaposi sarcoma-associated herpesvirus (KSHV), is a disease manifested mainly by dark purple skin and mouth nodules. Cancer care studies showed that co-infection of KSHV and human immunodeficiency virus (HIV) was able to increase the patients' survival, but the underlying mechanisms are still elusive. METHODS To understand the mechanism underlying the prolonged survival in KSHV-HIV co-infected patients, we performed microarray analysis on RNA extracted from biopsies from KS tumors and adjacent healthy tissues in four KS patients. Subsequently, we performed hierarchical clustering, gene ontology (GO) and ingenuity pathway analysis. We then characterized the roles of tight junction protein claudin-2 in the endothelial barrier function. RESULTS Three hundred and forty-three differentially expressed genes were identified, of which 246 genes exhibited significantly increased expression in the tumor compared to the adjacent healthy tissue and 97 genes showed downregulated expression, including claudin-2. Knockdown of claudin-2 in cultured endothelial cells enhances barrier function by altering the charge selectivity, but not the size selectivity. CONCLUSION Claudin-2 expression is decreased in KS tumors from patients co-infected with KSHV and HIV. Decreased claudin-2 enhances endothelial barrier function and may play a role in the prolonged survival of patients with KSHV and HIV co-infection.
Collapse
Affiliation(s)
- Xiaohua Tan
- School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | | | | | | | | | | |
Collapse
|
25
|
Chiu MH, Liu HS, Wu YH, Shen MR, Chou CY. SPAK mediates KCC3-enhanced cervical cancer tumorigenesis. FEBS J 2014; 281:2353-65. [DOI: 10.1111/febs.12787] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 02/17/2014] [Accepted: 03/18/2014] [Indexed: 11/28/2022]
Affiliation(s)
- Min-Hsi Chiu
- Institute of Basic Medical Sciences; National Cheng Kung University; Tainan Taiwan
| | - Hsiao-Sheng Liu
- Department of Microbiology and Immunology; National Cheng Kung University; Tainan Taiwan
| | - Yi-Hui Wu
- Department of Obstetrics and Gynecology; College of Medicine; National Cheng Kung University and Hospital; Tainan Taiwan
| | - Meng-Ru Shen
- Department of Obstetrics and Gynecology; College of Medicine; National Cheng Kung University and Hospital; Tainan Taiwan
- Department of Pharmacology; National Cheng Kung University; Tainan Taiwan
| | - Cheng-Yang Chou
- Department of Obstetrics and Gynecology; College of Medicine; National Cheng Kung University and Hospital; Tainan Taiwan
| |
Collapse
|
26
|
Centanni M, Turroni S, Consolandi C, Rampelli S, Peano C, Severgnini M, Biagi E, Caredda G, De Bellis G, Brigidi P, Candela M. The enterocyte-associated intestinal microbiota of breast-fed infants and adults responds differently to a TNF-α-mediated pro-inflammatory stimulus. PLoS One 2013; 8:e81762. [PMID: 24303069 PMCID: PMC3841132 DOI: 10.1371/journal.pone.0081762] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 10/16/2013] [Indexed: 12/20/2022] Open
Abstract
Co-evolved as an integral component of our immune system, the gut microbiota provides specific immunological services at different ages, supporting the immune education during our infancy and sustaining a well-balanced immunological homeostasis during the course of our life. In order to figure out whether this involves differences in the microbial groups primarily interacting with the host immune system, we developed a non-invasive HT29 cell-based minimal model to fingerprint the enterocyte-associated microbiota fraction in infants and adults. After depicting the fecal microbial community of 12 breast-fed infants and 6 adults by 16S rDNA amplicon pools 454 pyrosequencing, their respective HT29 cell-associated gut microbiota fractions were characterized by the universal phylogenetic array platform HTF-Microbi.Array, both in the presence and absence of a tumor necrosis factor-alpha (TNF-α)-mediated pro-inflammatory stimulus. Our data revealed remarkable differences between the enterocyte-associated microbiota fractions in breast-fed infants and adults, being dominated by Bifidobacterium and Enterobacteriaceae the first and Bacteroides-Prevotella and Clostridium clusters IV and XIVa the second. While in adults TNF-α resulted in a profound impairment of the structure of the enterocyte-associated microbiota fraction, in infants it remained unaffected. Differently from the adult-type gut microbial community, the infant-type microbiota is structured to cope with inflammation, being co-evolved to prime the early immune response by means of transient inflammatory signals from gut microorganisms.
Collapse
Affiliation(s)
- Manuela Centanni
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Silvia Turroni
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Clarissa Consolandi
- Institute of Biomedical Technologies - Italian National Research Council, Milan, Italy
| | - Simone Rampelli
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Clelia Peano
- Institute of Biomedical Technologies - Italian National Research Council, Milan, Italy
| | - Marco Severgnini
- Institute of Biomedical Technologies - Italian National Research Council, Milan, Italy
| | - Elena Biagi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Giada Caredda
- Institute of Biomedical Technologies - Italian National Research Council, Milan, Italy
| | - Gianluca De Bellis
- Institute of Biomedical Technologies - Italian National Research Council, Milan, Italy
| | - Patrizia Brigidi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Marco Candela
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
- * E-mail:
| |
Collapse
|
27
|
Zhang Y, Viennois E, Xiao B, Baker MT, Yang S, Okoro I, Yan Y. Knockout of Ste20-like proline/alanine-rich kinase (SPAK) attenuates intestinal inflammation in mice. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 182:1617-28. [PMID: 23499375 DOI: 10.1016/j.ajpath.2013.01.028] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 01/16/2013] [Accepted: 01/18/2013] [Indexed: 01/08/2023]
Abstract
Inflammatory bowel diseases are characterized by epithelial barrier disruption and alterations in immune regulation. Ste20-like proline/alanine-rich kinase (SPAK) plays a role in intestinal inflammation, but the underlying mechanisms need to be defined. Herein, SPAK knockout (KO) C57BL/6 mice exhibited significant increases in intestinal transepithelial resistance, a marked decrease in paracellular permeability to fluorescence isothiocyanate-dextran, and altered apical side tight junction sodium ion selectivity, compared with wild-type mice. Furthermore, the expression of junction protein, claudin-2, decreased. In contrast, expressions of occludin, E-cadherin, β-catenin, and claudin-5 increased significantly, whereas no obvious change of claudin-1, claudin-4, zonula occludens protein 1, and zonula occludens protein 2 expressions was observed. In murine models of colitis induced by dextran sulfate sodium and trinitrobenzene sulfuric acid, KO mice were more tolerant than wild-type mice, as demonstrated by colonoscopy features, histological characteristics, and myeloperoxidase activities. Consistent with these findings, KO mice showed increased IL-10 levels and decreased proinflammatory cytokine secretion, ameliorated bacterial translocation on treatment with dextran sulfate sodium, and regulation of with no lysine (WNK) kinase activity. Together, these features may reduce epithelial permeability. In conclusion, SPAK deficiency increases intestinal innate immune homeostasis, which is important for control or attenuation of pathological responses in inflammatory bowel diseases.
Collapse
Affiliation(s)
- Yuchen Zhang
- Department of Biology, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Gagnon KB, Delpire E. Molecular physiology of SPAK and OSR1: two Ste20-related protein kinases regulating ion transport. Physiol Rev 2013; 92:1577-617. [PMID: 23073627 DOI: 10.1152/physrev.00009.2012] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
SPAK (Ste20-related proline alanine rich kinase) and OSR1 (oxidative stress responsive kinase) are members of the germinal center kinase VI subfamily of the mammalian Ste20 (Sterile20)-related protein kinase family. Although there are 30 enzymes in this protein kinase family, their conservation across the fungi, plant, and animal kingdom confirms their evolutionary importance. Already, a large volume of work has accumulated on the tissue distribution, binding partners, signaling cascades, and physiological roles of mammalian SPAK and OSR1 in multiple organ systems. After reviewing this basic information, we will examine newer studies that demonstrate the pathophysiological consequences to SPAK and/or OSR1 disruption, discuss the development and analysis of genetically engineered mouse models, and address the possible role these serine/threonine kinases might have in cancer proliferation and migration.
Collapse
Affiliation(s)
- Kenneth B Gagnon
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-2520, USA
| | | |
Collapse
|
29
|
Jog SP, Paul S, Dansithong W, Tring S, Comai L, Reddy S. RNA splicing is responsive to MBNL1 dose. PLoS One 2012; 7:e48825. [PMID: 23166594 PMCID: PMC3499511 DOI: 10.1371/journal.pone.0048825] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Accepted: 10/03/2012] [Indexed: 11/24/2022] Open
Abstract
Myotonic dystrophy (DM1) is a highly variable, multi-system disorder resulting from the expansion of an untranslated CTG tract in DMPK. In DM1 expanded CUG repeat RNAs form hairpin secondary structures that bind and aberrantly sequester the RNA splice regulator, MBNL1. RNA splice defects resulting as a consequence of MBNL1 depletion have been shown to play a key role in the development of DM1 pathology. In patient populations, both the number and severity of DM1 symptoms increase broadly as a function of CTG tract length. However significant variability in the DM1 phenotype is observed in patients encoding similar CTG repeat numbers. Here we demonstrate that a gradual decrease in MBNL1 levels results both in the expansion of the repertoire of splice defects and an increase in the severity of the splice alterations. Thus, MBNL1 loss does not have an all or none outcome but rather shows a graded effect on the number and severity of the ensuing splice defects. Our results suggest that once a critical threshold is reached, relatively small dose variations of free MBNL1 levels, which may reflect modest changes in the size of the CUG tract or the extent of hairpin secondary structure formation, can significantly alter the number and severity of splice abnormalities and thus contribute to the phenotype variability observed in DM1 patients.
Collapse
Affiliation(s)
- Sonali P Jog
- Department of Biochemistry and Molecular Biology, Institute for Genetic Medicine, University of Southern California, Los Angeles, California, USA
| | | | | | | | | | | |
Collapse
|
30
|
Tumor necrosis factor alpha modulates the dynamics of the plasminogen-mediated early interaction between Bifidobacterium animalis subsp. lactis and human enterocytes. Appl Environ Microbiol 2012; 78:2465-9. [PMID: 22287006 DOI: 10.1128/aem.07883-11] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The capacity to intervene with the host plasminogen system has recently been considered an important component in the interaction process between Bifidobacterium animalis subsp. lactis and the human host. However, its significance in the bifidobacterial microecology within the human gastrointestinal tract is still an open question. Here we demonstrate that human plasminogen favors the B. animalis subsp. lactis BI07 adhesion to HT29 cells. Prompting the HT29 cell capacity to activate plasminogen, tumor necrosis factor alpha (TNF-α) modulated the plasminogen-mediated bacterium-enterocyte interaction, reducing the bacterial adhesion to the enterocytes and enhancing migration to the luminal compartment.
Collapse
|
31
|
Yan Y, Laroui H, Ingersoll SA, Ayyadurai S, Charania M, Yang S, Dalmasso G, Obertone TS, Nguyen H, Sitaraman SV, Merlin D. Overexpression of Ste20-related proline/alanine-rich kinase exacerbates experimental colitis in mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 187:1496-505. [PMID: 21705622 PMCID: PMC3140558 DOI: 10.4049/jimmunol.1002910] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Inflammatory bowel disease, mainly Crohn's disease and ulcerative colitis, are characterized by epithelial barrier disruption and altered immune regulation. Colonic Ste20-like proline/alanine-rich kinase (SPAK) plays a role in intestinal inflammation, but its underlying mechanisms need to be defined. Both SPAK-transfected Caco2-BBE cells and villin-SPAK transgenic (TG) FVB/6 mice exhibited loss of intestinal barrier function. Further studies demonstrated that SPAK significantly increased paracellular intestinal permeability to FITC-dextran. In vivo studies using the mouse models of colitis induced by dextran sulfate sodium (DSS) and trinitrobenzene sulfonic acid showed that TG FVB/6 mice were more susceptible to DSS and trinitrobenzene sulfonic acid treatment than wild-type FVB/6 mice, as demonstrated by clinical and histological characteristics and enzymatic activities. Consistent with this notion, we found that SPAK increased intestinal epithelial permeability, which likely facilitated the production of inflammatory cytokines in vitro and in vivo, aggravated bacterial translocation in TG mice under DSS treatment, and consequently established a context favorable for the triggering of intestinal inflammation cascades. In conclusion, overexpression of SPAK inhibits maintenance of intestinal mucosal innate immune homeostasis, which makes regulation of SPAK important to attenuate pathological responses in inflammatory bowel disease.
Collapse
Affiliation(s)
- Yutao Yan
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Diesel B, Ripoche N, Risch RT, Tierling S, Walter J, Kiemer AK. Inflammation-induced up-regulation of TLR2 expression in human endothelial cells is independent of differential methylation in the TLR2 promoter CpG island. Innate Immun 2011; 18:112-23. [PMID: 21768203 DOI: 10.1177/1753425910394888] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Toll-like receptors play an important role in endothelial inflammation; however, little is known on the mechanisms regulating their expression. Differential promoter DNA methylation is an increasingly recognized mechanism that determines a switch between gene silencing and gene transcription. We hypothesized that epigenetic mechanisms are involved in the regulation of endothelial TLR2 expression because of the localization of the TLR2 promoter on a CpG-island. Resting human umbilical vein endothelial cells (HUVECs) displayed rather low TLR2 mRNA expression, while a strong expression increase occurred under inflammatory conditions. We examined the TLR2 promoter methylation pattern in resting HUVECs and compared it to cells treated either with the inflammatory cytokine TNF-α or the DNA-demethylating agent 5-azacytidine. DNA bisulfite conversion was followed by either genomic sequencing or single nucleotide primer extension (SNuPE) HPLC. Results of both techniques showed a low- or non-methylated TLR2 promoter in resting HUVECs and no alteration of the methylation pattern under inflammatory conditions. Whereas 5-azacytidine significantly increased the mRNA expression of the epigenetically regulated gene H19, TLR2 expression was not affected. Taken together, employing different methodological approaches, our data show no implication of methylation pattern changes in inflammatory induction of TLR2 expression in human endothelial cells.
Collapse
Affiliation(s)
- Britta Diesel
- Department of Pharmaceutical Biology, Saarland University, Saarbrücken, Germany
| | | | | | | | | | | |
Collapse
|
33
|
Hashimoto K, Oreffo ROC, Gibson MB, Goldring MB, Roach HI. DNA demethylation at specific CpG sites in the IL1B promoter in response to inflammatory cytokines in human articular chondrocytes. ACTA ACUST UNITED AC 2010; 60:3303-13. [PMID: 19877066 DOI: 10.1002/art.24882] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE To determine whether changes in the DNA methylation status in the promoter region of the gene encoding interleukin-1beta (IL-1beta) account for expression of IL1B messenger RNA (mRNA) after long-term treatment of human articular chondrocytes with inflammatory cytokines. METHODS IL-1beta, tumor necrosis factor alpha (TNFalpha) plus oncostatin M (OSM), or 5-azadeoxycytidine (5-aza-dC) was added twice weekly for 4-5 weeks to primary cultures of normal human articular chondrocytes derived from the femoral head cartilage of patients with a fracture of the femoral neck. Expression of MMP13, IL1B, TNFA, and DNMT1 was determined by SYBR Green-based quantitative reverse transcription-polymerase chain reaction (RT-PCR) analysis of genomic DNA and total RNA extracted from the same sample before and after culture. Bisulfite modification was used to identify which CpG sites in the IL1B promoter showed differential methylation between IL1B-expressing and IL1B-nonexpressing cells. The percentages of cells that were methylated at that critical CpG site (-299 bp) were quantified by a method that depended on methylation-sensitive restriction enzymes and real-time RT-PCR. Secretion of IL-1beta into the culture media was assessed by enzyme-linked immunosorbent assay. RESULTS Healthy chondrocytes did not express IL1B mRNA, but the levels were increased 5-fold by treatment with 5-aza-dC and were increased 100-1,000-fold by treatment with TNFalpha/OSM. The percentage CpG methylation was decreased by 5-aza-dC treatment but was reduced considerably more by IL-1beta and was almost abolished by TNFalpha/OSM. The mRNA was translated into protein in cytokine-treated chondrocytes. CONCLUSION These novel findings indicate that inflammatory cytokines can change the DNA methylation status at key CpG sites, resulting in long-term induction of IL1B in human articular chondrocytes.
Collapse
|
34
|
Ste20-related proline/alanine-rich kinase (SPAK) regulated transcriptionally by hyperosmolarity is involved in intestinal barrier function. PLoS One 2009; 4:e5049. [PMID: 19343169 PMCID: PMC2660421 DOI: 10.1371/journal.pone.0005049] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2009] [Accepted: 02/06/2009] [Indexed: 01/01/2023] Open
Abstract
The Ste20-related protein proline/alanine-rich kinase (SPAK) plays important roles in cellular functions such as cell differentiation and regulation of chloride transport, but its roles in pathogenesis of intestinal inflammation remain largely unknown. Here we report significantly increased SPAK expression levels in hyperosmotic environments, such as mucosal biopsy samples from patients with Crohn's disease, as well as colon tissues of C57BL/6 mice and Caco2-BBE cells treated with hyperosmotic medium. NF-kappaB and Sp1-binding sites in the SPAK TATA-less promoter are essential for SPAK mRNA transcription. Hyperosmolarity increases the ability of NF-kappaB and Sp1 to bind to their binding sites. Knock-down of either NF-kappaB or Sp1 by siRNA reduces the hyperosmolarity-induced SPAK expression levels. Furthermore, expression of NF-kappaB, but not Sp1, was upregulated by hyperosmolarity in vivo and in vitro. Nuclear run-on assays showed that hyperosmolarity increases SPAK expression levels at the transcriptional level, without affecting SPAK mRNA stability. Knockdown of SPAK expression by siRNA or overexpression of SPAK in cells and transgenic mice shows that SPAK is involved in intestinal permeability in vitro and in vivo. Together, our data suggest that SPAK, the transcription of which is regulated by hyperosmolarity, plays an important role in epithelial barrier function.
Collapse
|
35
|
Teng X, Xu LF, Zhou P, Sun HW, Sun M. Effects of trefoil peptide 3 on expression of TNF-alpha, TLR4, and NF-kappaB in trinitrobenzene sulphonic acid induced colitis mice. Inflammation 2009; 32:120-129. [PMID: 19238529 DOI: 10.1007/s10753-009-9110-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The trefoil factor (TFF) peptides are major secretory products of mucus cells of the gastrointestinal tract. There were evidences that administration of recombinant human TFF3 is effective in treatment of models of colitis, but the mechanism of the effects of rTFF3 is not fully understood. The main aims of this study is to evaluate effects of intraperitoneal injection recombinant human TFF3 on the expression of tumour necrosis factor alpha (TNF-alpha), toll-like receptor 4(TLR4), and nuclear factor kappaB (NF-kappaB) in trinitrobenzene sulphonic acid (TNBS) induced colitis mice. Distal colitis was induced in BALB/C mice by intracolonic administration of TNBS in ethanol. Treated with administration rhTFF3 for treatment group(5 mg/ml; approximately 0.5 mg/mouse), and normal saline for control for 5 consecutive days. Colonic damage score, tissue myeloperoxidase (MPO) activity, TLR4, NF-kappaB mRNA expression, and tissue TNF-alpha, TLR4, NF-kappaB production were determined, respectively. Once daily application of hTFF3 for 5 days after TNBS/ethanol had been injected, both microscopic and macroscopic injury and inflammatory index had been reduced compared with controls. In addition, decreased tissue TNF-alpha, TLR4, NF-kappaB production, and TLR4, NF-kappaB mRNA expression had been found. This study has shown that hTFF3 may have therapeutic potential in the treatment of inflammatory bowel disease, and one of the mechanisms may related to inhibit the TLR4/NF-kappaB signaling pathways.
Collapse
Affiliation(s)
- Xu Teng
- Department of Pediatric Gastroenterology, Shengjing Hospital Of China Medical University, Shenyang, 110004, Liaoning, China
| | | | | | | | | |
Collapse
|