1
|
Snyder JD, Yoon TW, Lee S, Halder P, Fitzpatrick EA, Yi AK. Protein kinase D1 in myeloid lineage cells contributes to the accumulation of CXCR3 +CCR6 + nonconventional Th1 cells in the lungs and potentiates hypersensitivity pneumonitis caused by S. rectivirgula. Front Immunol 2024; 15:1403155. [PMID: 39464896 PMCID: PMC11502317 DOI: 10.3389/fimmu.2024.1403155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 09/18/2024] [Indexed: 10/29/2024] Open
Abstract
Introduction Hypersensitivity pneumonitis (HP) is an extrinsic allergic alveolitis characterized by inflammation of the interstitium, bronchioles, and alveoli of the lung that leads to granuloma formation. We previously found that activation of protein kinase D1 (PKD1) in the lungs following exposures to Saccharopolyspora rectivirgula contributes to the acute pulmonary inflammation, IL-17A expression in the lungs, and development of HP. In the present study, we investigated whether PKD1 in myeloid-lineage cells affects the pathogenic course of the S. rectivirgula-induced HP. Methods Mice were exposed intranasally to S. rectivirgula once or 3 times/week for 3 weeks. The protein and mRNA expression levels of cytokines/chemokines were detected by enzyme-linked immunosorbent assay and quantitative real-time PCR, respectively. Flow cytometry was used to detect the different types of immune cells and the levels of surface proteins. Lung tissue sections were stained with hematoxylin and eosin, digital images were captured, and immune cells influx into the interstitial lung tissue were detected. Results Compared to control PKD1-sufficient mice, mice with PKD1 deficiency in myeloid-lineage cells (PKD1mKO) showed significantly suppressed expression of TNFα, IFNγ, IL-6, CCL2, CCL3, CCL4, CXCL1, CXCL2, and CXCL10 and neutrophilic alveolitis after single intranasal exposure to S. rectivirgula. Substantially reduced levels of alveolitis and granuloma formation were observed in the PKD1mKO mice repeatedly exposed to S. rectivirgula for 3 weeks. In addition, expression levels of the Th1/Th17 polarizing cytokines and chemokines such as IFNγ, IL-17A, CXCL9, CXCL10, CXCL11, and CCL20 in lungs were significantly reduced in the PKD1mKO mice repeatedly exposed to S. rectivirgula. Moreover, accumulation of CXCR3+CCR6+ nonconventional Th1 in the lungs were significantly reduced in PKD1mKO mice repeatedly exposed to S. rectivirgula. Discussion Our results demonstrate that PKD1 in myeloid-lineage cells plays an essential role in the development and progress of HP caused by repeated exposure to S. rectivirgula by contributing Th1/Th17 polarizing proinflammatory responses, alveolitis, and accumulation of pathogenic nonconventional Th1 cells in the lungs.
Collapse
Affiliation(s)
- John D. Snyder
- Integrated Biomedical Science Graduate Program, The University of Tennessee Health Science Center, Memphis, TN, United States
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Tae Won Yoon
- Integrated Biomedical Science Graduate Program, The University of Tennessee Health Science Center, Memphis, TN, United States
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Sangmin Lee
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Priyanka Halder
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Elizabeth Ann Fitzpatrick
- Integrated Biomedical Science Graduate Program, The University of Tennessee Health Science Center, Memphis, TN, United States
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Ae-Kyung Yi
- Integrated Biomedical Science Graduate Program, The University of Tennessee Health Science Center, Memphis, TN, United States
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
2
|
Clites BL, Frohock B, Koury EJ, Andersen EC, Pierce JT. Natural variation in protein kinase D modifies alcohol sensitivity in Caenorhabditis elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.09.598102. [PMID: 38895441 PMCID: PMC11185769 DOI: 10.1101/2024.06.09.598102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Differences in naïve alcohol sensitivity between individuals are a strong predictor of later life alcohol use disorders (AUD). However, the genetic bases for alcohol sensitivity (beyond ethanol metabolism) and pharmacological approaches to modulate alcohol sensitivity remain poorly understood. We used a high-throughput behavioral screen to measure acute behavioral sensitivity to alcohol, a model of intoxication, in a genetically diverse set of over 150 wild strains of the nematode Caenorhabditis elegans. We performed a genome-wide association study to identify loci that underlie natural variation in alcohol sensitivity. We identified five quantitative trait loci (QTL) and further show that variants in the C. elegans ortholog of protein kinase D, dkf-2, likely underlie the chromosome V QTL. We found that resistance to intoxication was conferred by dkf-2 loss-of-function mutations as well as partly by a PKD inhibitor in a dkf-2-dependent manner. Protein kinase D might represent a conserved, druggable target to modify alcohol sensitivity with application towards AUD.
Collapse
Affiliation(s)
- Benjamin L Clites
- Waggoner Center for Alcohol & Addiction Research, Center for Learning and Memory, Department of Neuroscience, University of Texas at Austin, Austin TX
| | - Brooke Frohock
- Waggoner Center for Alcohol & Addiction Research, Center for Learning and Memory, Department of Neuroscience, University of Texas at Austin, Austin TX
| | - Emily J Koury
- Department of Biology, Johns Hopkins University, Baltimore MD
| | - Erik C Andersen
- Department of Biology, Johns Hopkins University, Baltimore MD
| | - Jonathan T Pierce
- Waggoner Center for Alcohol & Addiction Research, Center for Learning and Memory, Department of Neuroscience, University of Texas at Austin, Austin TX
| |
Collapse
|
3
|
Wu L, Hu J, Yi X, Lv J, Yao J, Tang W, Zhang S, Wan M. Gut microbiota interacts with inflammatory responses in acute pancreatitis. Therap Adv Gastroenterol 2023; 16:17562848231202133. [PMID: 37829561 PMCID: PMC10566291 DOI: 10.1177/17562848231202133] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 09/01/2023] [Indexed: 10/14/2023] Open
Abstract
Acute pancreatitis (AP) is one of the most common acute abdominal conditions, and its incidence has been increasing for years. Approximately 15-20% of patients develop severe AP (SAP), which is complicated by critical inflammatory injury and intestinal dysfunction. AP-associated inflammation can lead to the gut barrier and function damage, causing dysbacteriosis and facilitating intestinal microbiota migration. Pancreatic exocrine deficiency and decreased levels of antimicrobial peptides in AP can also lead to abnormal growth of intestinal bacteria. Meanwhile, intestinal microbiota migration influences the pancreatic microenvironment and affects the severity of AP, which, in turn, exacerbates the systemic inflammatory response. Thus, the interaction between the gut microbiota (GM) and the inflammatory response may be a key pathogenic feature of SAP. Treating either of these factors or breaking their interaction may offer some benefits for SAP treatment. In this review, we discuss the mechanisms of interaction of the GM and inflammation in AP and factors that can deteriorate or even cure both, including some traditional Chinese medicine treatments, to provide new methods for studying AP pathogenesis and developing therapies.
Collapse
Affiliation(s)
- Linjun Wu
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
- Hospital of Chinese Traditional Medicine of Leshan, Leshan, China
| | - Jing Hu
- Department of Integrated Traditional Chinese and Western Medicine, West China
- Hospital, Sichuan University, Chengdu, China
- Hospital of Chinese Traditional Medicine of Leshan, Leshan, China
| | - Xiaolin Yi
- Department of Integrated Traditional Chinese and Western Medicine, West China
- Hospital, Sichuan University, Chengdu, China
- Intensive Care Unit, Suining Municipal Hospital of TCM, Suining, China
| | - Jianqin Lv
- Department of Integrated Traditional Chinese and Western Medicine, West China
- Hospital, Sichuan University, Chengdu, China
| | - Jiaqi Yao
- Department of Integrated Traditional Chinese and Western Medicine, West China
- Hospital, Sichuan University, Chengdu, China
| | - Wenfu Tang
- Department of Integrated Traditional Chinese and Western Medicine, West China
- Hospital, Sichuan University, Chengdu, China
| | - Shu Zhang
- Department of Emergency Medicine, Emergency Medical Laboratory, West China
- Hospital, Sichuan University, Guo Xue Road 37, Chengdu 610041, Sichuan, China
| | - Meihua Wan
- Department of Integrated Traditional Chinese and Western Medicine, West China
- Hospital, Sichuan University, Guo Xue Road 37, Chengdu 610041, China
| |
Collapse
|
4
|
Cui B, Liu Y, Chen J, Chen H, Feng Y, Zhang P. Small molecule inhibitor CRT0066101 inhibits cytokine storm syndrome in a mouse model of lung injury. Int Immunopharmacol 2023; 120:110240. [PMID: 37182445 PMCID: PMC10181585 DOI: 10.1016/j.intimp.2023.110240] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/08/2023] [Accepted: 04/22/2023] [Indexed: 05/16/2023]
Abstract
Pneumonia is an acute inflammation of the lungs induced by pathogenic microorganisms, immune damage, physical and chemical factors, and other factors, and the latest outbreak of novel coronavirus pneumonia is also an acute lung injury (ALI) induced by viral infection. However, there are currently no effective treatments for inflammatory cytokine storms in patients with ALI/acute respiratory distress syndrome (ARDS). Protein kinase D (PKD) is a highly active kinase that has been shown to be associated with the production of inflammatory cytokines. Therefore, small-molecule compounds that inhibit PKD may be potential drugs for the treatment of ALI/ARDS. In the present study, we evaluated the ability of the small-molecule inhibitor CRT0066101 to attenuate lipopolysaccharide (LPS)-induced inflammatory cytokine production through in vitro cell experiments and a mouse pneumonia model. We found that CRT0066101 significantly reduced the protein and mRNA levels of LPS-induced cytokines (e.g., IL-6, TNF-α, and IL-1β). CRT0066101 inhibited MyD88 and TLR4 expression and reduced NF-κB, ERK, and JNK phosphorylation. CRT0066101 also reduced NLRP3 activation, inhibited the assembly of the inflammasome complex, and attenuated inflammatory cell infiltration and lung tissue damage. Taken together, our data indicate that CRT0066101 exerts anti-inflammatory effects on LPS-induced inflammation through the TLR4/MyD88 signaling pathway, suggesting that CRT0066101 may have therapeutic value in acute lung injury and other MyD88-dependent inflammatory diseases.
Collapse
Affiliation(s)
- Bomiao Cui
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, 14, Renmin South Road Section 3, Chengdu, Sichuan 610041, PR China
| | - Yiying Liu
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, 14, Renmin South Road Section 3, Chengdu, Sichuan 610041, PR China
| | - Jiao Chen
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, 14, Renmin South Road Section 3, Chengdu, Sichuan 610041, PR China
| | - Hongli Chen
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, 14, Renmin South Road Section 3, Chengdu, Sichuan 610041, PR China
| | - Yun Feng
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, 14, Renmin South Road Section 3, Chengdu, Sichuan 610041, PR China
| | - Ping Zhang
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, 14, Renmin South Road Section 3, Chengdu, Sichuan 610041, PR China.
| |
Collapse
|
5
|
Burciaga SD, Saavedra F, Fischer L, Johnstone K, Jensen ED. Protein kinase D3 conditional knockout impairs osteoclast formation and increases trabecular bone volume in male mice. Bone 2023; 172:116759. [PMID: 37044359 DOI: 10.1016/j.bone.2023.116759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/15/2023] [Accepted: 04/04/2023] [Indexed: 04/14/2023]
Abstract
Studies using kinase inhibitors have shown that the protein kinase D (PRKD) family of serine/threonine kinases are required for formation and function of osteoclasts in culture. However, the involvement of individual protein kinase D genes and their in vivo significance to skeletal dynamics remains unclear. In the current study we present data indicating that protein kinase D3 is the primary form of PRKD expressed in osteoclasts. We hypothesized that loss of PRKD3 would impair osteoclast formation, thereby decreasing bone resorption and increasing bone mass. Conditional knockout (cKO) of Prkd3 using a murine Cre/Lox system driven by cFms-Cre revealed that its loss in osteoclast-lineage cells reduced osteoclast differentiation and resorptive function in culture. Examination of the Prkd3 cKO mice showed that bone parameters were unaffected in the femur at 4 weeks of age, but consistent with our hypothesis, Prkd3 conditional knockout resulted in 18 % increased trabecular bone mass in male mice at 12 weeks and a similar increase at 6 months. These effects were not observed in female mice. As a further test of our hypothesis, we asked if Prkd3 cKO could protect against bone loss in a ligature-induced periodontal disease model but did not see any reduction in bone destruction in this system. Together, our data indicate that PRKD3 promotes osteoclastogenesis both in vitro and in vivo.
Collapse
Affiliation(s)
- Samuel D Burciaga
- Department of Diagnostic & Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, MN 55455, USA
| | - Flavia Saavedra
- Department of Diagnostic & Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, MN 55455, USA
| | - Lori Fischer
- Department of Diagnostic & Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, MN 55455, USA
| | - Karen Johnstone
- Department of Diagnostic & Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, MN 55455, USA
| | - Eric D Jensen
- Department of Diagnostic & Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, MN 55455, USA.
| |
Collapse
|
6
|
Veazey JM, Wong GS, Eliseeva SI, Smyth TR, Chapman TJ, Lim K, Kim M, Georas SN. Protein kinase D3 promotes neutrophil migration during viral infection. Immunol Cell Biol 2023; 101:130-141. [PMID: 36318273 PMCID: PMC10112008 DOI: 10.1111/imcb.12603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/13/2022] [Accepted: 10/30/2022] [Indexed: 11/05/2022]
Abstract
Protein kinase D (PKD) is a serine/threonine kinase family with three isoforms (PKD1-3) that are expressed in most cells and implicated in a wide array of signaling pathways, including cell growth, differentiation, transcription, secretion, polarization and actin turnover. Despite growing interest in PKD, relatively little is known about the role of PKD in immune responses. We recently published that inhibiting PKD limits proinflammatory cytokine secretion and leukocyte accumulation in mouse models of viral infection, and that PKD3 is highly expressed in the murine lung and immune cell populations. Here we focus on the immune-related phenotypes of PKD3 knockout mice. We report that PKD3 is necessary for maximal neutrophil accumulation in the lung following challenge with inhaled polyinosinic:polycytidylic acid, a double-stranded RNA, as well as following influenza A virus infection. Using reciprocal bone marrow chimeras, we found that PKD3 is required in the hematopoietic compartment for optimal neutrophil migration to the lung. Ex vivo transwell and chemokinesis assays confirmed that PKD3-/- neutrophils possess an intrinsic motility defect, partly because of reduced surface expression of CD18, which is critical for leukocyte migration. Finally, the peak of neutrophilia was significantly reduced in PKD3-/- mice after lethal influenza A virus infection. Together, these results demonstrate that PKD3 has an essential, and nonredundant, role in promoting neutrophil recruitment to the lung. A better understanding of the isoform-specific and cell type-specific activities of PKD has the potential to lead to novel therapeutics for respiratory illnesses.
Collapse
Affiliation(s)
- Janelle M Veazey
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA
| | - Gordon S Wong
- Department of Medicine, Yale New Haven Health, Greenwich Hospital, Greenwich, CT, USA
- Department of Medicine, Pulmonary and Critical Care, University of Rochester, Rochester, NY, USA
| | - Sophia I Eliseeva
- Department of Medicine, Pulmonary and Critical Care, University of Rochester, Rochester, NY, USA
| | - Timothy R Smyth
- Department of Toxicology, University of North Caroline, Chapel Hill, NC, USA
- Department of Environmental Medicine, University of Rochester, Rochester, NY, USA
| | - Timothy J Chapman
- Department of Medicine, Pulmonary and Critical Care, University of Rochester, Rochester, NY, USA
- Merck, Kenilworth, NJ, USA
| | - Kihong Lim
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA
| | - Minsoo Kim
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA
| | - Steve N Georas
- Department of Medicine, Pulmonary and Critical Care, University of Rochester, Rochester, NY, USA
| |
Collapse
|
7
|
Chen L, Zhao J, Chao Y, Roy A, Guo W, Qian J, Xu W, Domsic RT, Lafyatis R, Lu B, Deng F, Wang QJ. Loss of Protein Kinase D2 Activity Protects Against Bleomycin-Induced Dermal Fibrosis in Mice. J Transl Med 2023; 103:100018. [PMID: 37039152 PMCID: PMC10507682 DOI: 10.1016/j.labinv.2022.100018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 08/22/2022] [Accepted: 09/20/2022] [Indexed: 01/11/2023] Open
Abstract
Protein kinase D (PKD) has been linked to inflammatory responses in various pathologic conditions; however, its role in inflammation-induced dermal fibrosis has not been evaluated. In this study, we aimed to investigate the roles and mechanisms of protein kinase D2 (PKD2) in inflammation-induced dermal fibrosis and evaluate the therapeutic potential of PKD inhibitors in this disease. Using homozygous kinase-dead PKD2 knock-in (KI) mice, we examined whether genetic ablation or pharmacologic inhibition of PKD2 activity affected dermal inflammation and fibrosis in a bleomycin (BLM)-induced skin fibrosis model. Our data showed that dermal thickness and collagen fibers were significantly reduced in BLM-treated PKD2 KI mice compared with that in wild-type mice, and so was the expression of α-smooth muscle actin and collagens and the mRNA levels of transforming growth factor-β1 and interleukin-6 in the KI mice. Corroboratively, pharmacologic inhibition of PKD by CRT0066101 also significantly blocked BLM-induced dermal fibrosis and reduced α-smooth muscle actin, collagen, and interleukin-6 expression. Further analyses indicated that loss of PKD2 activity significantly blocked BLM-induced infiltration of monocytes/macrophages and neutrophils in the dermis. Moreover, using bone marrow-derived macrophages, we demonstrated that PKD activity was required for cytokine production and migration of macrophages. We have further identified Akt as a major downstream target of PKD2 in the early inflammatory phase of the fibrotic process. Taken together, our findings indicate that PKD2 promotes dermal fibrosis via regulating immune cell infiltration, cytokine production, and downstream activation of Akt in lesional skin, and targeted inhibition of PKD2 may benefit the treatment of this condition.
Collapse
Affiliation(s)
- Liping Chen
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jinjun Zhao
- Department of Rheumatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yapeng Chao
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Adhiraj Roy
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, India
| | - Wenjing Guo
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jiabi Qian
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Wanfu Xu
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Robyn T Domsic
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Robert Lafyatis
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Binfeng Lu
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Fan Deng
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Q Jane Wang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
8
|
Senturk ZN, Akdag I, Deniz B, Sayi-Yazgan A. Pancreatic cancer: Emerging field of regulatory B-cell-targeted immunotherapies. Front Immunol 2023; 14:1152551. [PMID: 37033931 PMCID: PMC10076755 DOI: 10.3389/fimmu.2023.1152551] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/10/2023] [Indexed: 04/11/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), the most common type of pancreatic cancer, is characterized by a high mortality rate and poor prognosis. Current treatments for PDAC, are ineffective due to a prominent immunosuppressive PDAC tumor microenvironment (TME). Although B lymphocytes are highly infiltrated into PDAC, the importance of B lymphocytes in tumorigenesis is largely neglected. B cells play a dual role in the PDAC tumor microenvironment, acting as either anti-tumorigenic or pro-tumorigenic depending on where they are localized. Tumor-infiltrating B cells, which reside in ectopic lymph nodes, namely tertiary lymphoid structures (TLS), produce anti-tumor antibodies and present tumor antigens to T cells to contribute to cancer immunosurveillance. Alternatively, regulatory B cells (Bregs), dispersed inside the TME, contribute to the dampening of anti-tumor immune responses by secreting anti-inflammatory cytokines (IL-10 and IL-35), which promote tumor growth and metastasis. Determining the role of Bregs in the PDAC microenvironment is thus becoming increasingly attractive for developing novel immunotherapeutic approaches. In this minireview, we shed light on the emerging role of B cells in PDAC development and progression, with an emphasis on regulatory B cells (Bregs). Furthermore, we discussed the potential link of Bregs to immunotherapies in PDAC. These current findings will help us in understanding the full potential of B cells in immunotherapy.
Collapse
|
9
|
Najmanová L, Vídeňská P, Cahová M. Healthy microbiome – a mere idea or a sound concept? Physiol Res 2022. [PMID: 36426891 DOI: 10.33549/physiolres.934967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Hundreds of studies in last decades have aimed to compare the microbiome of patients suffering from diverse diseases with that of healthy controls. The microbiome-related component was additionally identified in pathophysiology of many diseases formerly considered to depend only on the host physiology. This, however, opens important questions like: “What is the healthy microbiome?” or “Is it possible to define it unequivocally?”. In this review, we describe the main hindrances complicating the definition of “healthy microbiome” in terms of microbiota composition. We discuss the human microbiome from the perspective of classical ecology and we advocate for the shift from the stress on microbiota composition to the functions that microbiome ensures for the host. Finally, we propose to leave the concept of ideal healthy microbiome and replace it by focus on microbiome advantageous for the host, which always depends on the specific context like the age, genetics, dietary habits, body site or physiological state.
Collapse
Affiliation(s)
| | | | - M Cahová
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| |
Collapse
|
10
|
Pediatric hypersensitivity pneumonitis: literature update and proposal of a diagnostic algorithm. Ital J Pediatr 2022; 48:51. [PMID: 35346317 PMCID: PMC8962565 DOI: 10.1186/s13052-022-01239-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 03/09/2022] [Indexed: 11/10/2022] Open
Abstract
Hypersensitivity pneumonitis (HP) is a rare disease in childhood with the prevalence of 4 cases per 1 million children and an incidence of 2 cases per year. The average age of diagnosis at pediatric age is approximately 10 years. The pathogenesis of HP is characterized by an immunological reaction caused by recurrent exposure to triggering environmental agents (mostly bird antigens in children). The clinical picture of HP is complex and variable in children, often presenting in subacute forms with cough and exertion dyspnea. A diagnosis of HP should be considered in patients with an identified exposure to a triggering antigen, respiratory symptoms, and radiologic signs of interstitial lung disease. Blood tests and pulmonary function tests (PFT) support the diagnosis. Bronchoscopy (with bronchoalveolar lavage and tissue biopsy) may be needed in unclear cases. Antigen provocation test is rarely required. Of note, the persistence of symptoms despite various treatment regimens may support HP diagnosis. The avoidance of single/multiple triggers is crucial for effective treatment. No evidence- based guidelines for treatment are available; in particular, the role of systemic glucocorticoids in children is unclear. With adequate antigen avoidance, the prognosis in children with HP is generally favorable.
Collapse
|
11
|
Vo TTT, Huang HW, Wee Y, Feng SW, Cheng HC, Tuan VP, Lee IT. Surfactin reduces particulate matter-induced VCAM-1-dependent monocyte adhesion in human gingival fibroblasts by increasing Nrf2-dependent HO-1 expression. J Periodontal Res 2021; 57:115-130. [PMID: 34716926 DOI: 10.1111/jre.12944] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/19/2021] [Accepted: 10/13/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND OBJECTIVES The mechanisms of particulate matter (PM) toxicity involve the generation of ROS and upregulation of proinflammatory molecules. Nrf2 is a multifunctional cytoprotective transcription factor that regulates the expression of various antioxidant, anti-inflammatory, and detoxifying molecules, such as HO-1. As surfactin has potential to induce Nrf2 activation and HO-1 expression, this study aimed to investigate the anti-inflammatory effects of surfactin on PM-exposed human gingival fibroblasts (HGFs) and signaling pathways engaged by surfactin. MATERIALS AND METHODS Human gingival fibroblasts were challenged by PM with or without surfactin pretreatment. The expression of Nrf2, HO-1, VCAM-1, and other molecules was determined by western blot, real-time PCR, or ELISA. Human monocytic THP-1 cells labeled with fluorescent reagent were added to HGFs, and the cell adhesion was assessed. ROS generation and NADPH oxidase activity were also measured. The involvement of Nrf2/HO-1 and ROS signaling pathways was investigated by treating HGFs with specific pathway interventions, genetically or pharmacologically. One dose of surfactin was given to mice before PM treatment to explore its in vivo effect on VCAM-1 expression in gingival tissues. RESULTS Particulate matter led to VCAM-1-dependent monocyte adhesion in HGFs, which was regulated by PKCα/NADPH oxidase/ROS/STAT1/IL-6 pathway. Surfactin could attenuate monocyte adhesion by disrupting this VCAM-1-dependent pathway. Additionally, surfactin promoted Nrf2-dependent HO-1 expression in HGFs, mitigating VCAM-1 expression. PM-treated mice exhibited the lower expression of IL-6 and VCAM-1 in gingival tissues if they previously received surfactin. CONCLUSION Surfactin exerts anti-inflammatory effects against PM-induced inflammatory responses in HGFs by inhibiting VCAM-1-dependent pathway and inducing Nrf2/HO-1 axis.
Collapse
Affiliation(s)
- Thi Thuy Tien Vo
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Han Wei Huang
- Department of Dentistry, Taipei Medical University Hospital, Taipei, Taiwan
| | - Yinshen Wee
- Department of Pathology, University of Utah, Salt Lake City, Utah, USA
| | - Sheng-Wei Feng
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Dentistry, Taipei Medical University Hospital, Taipei, Taiwan
| | - Hsin-Chung Cheng
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Dentistry, Taipei Medical University Hospital, Taipei, Taiwan
| | - Vo Phuoc Tuan
- Endoscopy Department, Cho Ray Hospital, Ho Chi Minh City, Vietnam
| | - I-Ta Lee
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
12
|
Chandra V, McAllister F. Therapeutic potential of microbial modulation in pancreatic cancer. Gut 2021; 70:gutjnl-2019-319807. [PMID: 33906958 PMCID: PMC8292583 DOI: 10.1136/gutjnl-2019-319807] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 03/16/2021] [Accepted: 04/12/2021] [Indexed: 02/06/2023]
Affiliation(s)
- Vidhi Chandra
- Department of Clinical Cancer Prevention, Houston, Texas, USA
| | - Florencia McAllister
- Department of Clinical Cancer Prevention, Houston, Texas, USA
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
13
|
Zhang X, Connelly J, Chao Y, Wang QJ. Multifaceted Functions of Protein Kinase D in Pathological Processes and Human Diseases. Biomolecules 2021; 11:biom11030483. [PMID: 33807058 PMCID: PMC8005150 DOI: 10.3390/biom11030483] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/13/2021] [Accepted: 03/15/2021] [Indexed: 02/06/2023] Open
Abstract
Protein kinase D (PKD) is a family of serine/threonine protein kinases operating in the signaling network of the second messenger diacylglycerol. The three family members, PKD1, PKD2, and PKD3, are activated by a variety of extracellular stimuli and transduce cell signals affecting many aspects of basic cell functions including secretion, migration, proliferation, survival, angiogenesis, and immune response. Dysregulation of PKD in expression and activity has been detected in many human diseases. Further loss- or gain-of-function studies at cellular levels and in animal models provide strong support for crucial roles of PKD in many pathological conditions, including cancer, metabolic disorders, cardiac diseases, central nervous system disorders, inflammatory diseases, and immune dysregulation. Complexity in enzymatic regulation and function is evident as PKD isoforms may act differently in different biological systems and disease models, and understanding the molecular mechanisms underlying these differences and their biological significance in vivo is essential for the development of safer and more effective PKD-targeted therapies. In this review, to provide a global understanding of PKD function, we present an overview of the PKD family in several major human diseases with more focus on cancer-associated biological processes.
Collapse
|
14
|
Veazey JM, Eliseeva SI, Hillman SE, Stiles K, Smyth TR, Morrissey CE, Tillotson EJ, Topham DJ, Chapman TJ, Georas SN. Inhibiting Protein Kinase D Promotes Airway Epithelial Barrier Integrity in Mouse Models of Influenza A Virus Infection. Front Immunol 2020; 11:580401. [PMID: 33381112 PMCID: PMC7767883 DOI: 10.3389/fimmu.2020.580401] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/05/2020] [Indexed: 11/13/2022] Open
Abstract
Rationale Protein kinase D (PKD) is a serine/threonine kinase family that is involved in a wide array of signaling pathways. Although PKD has been implicated in immune responses, relatively little is known about the function of PKD in the lung or during viral infections. Objectives We investigated the hypothesis that PKD is involved in multiple aspects of host response to viral infection. Methods The selective PKD inhibitor CRT0010166 was administered to C57BL/6 mice prior to and during challenge with either inhaled double-stranded RNA or Influenza A Virus. PKD signaling pathways were investigated in human bronchial epithelial cells treated with CRT0010166, double-stranded RNA, and/or infected with Influenza A Virus. Measurements Total protein and albumin accumulation in the bronchoalveolar fluid was used to asses inside/out leak. Clearance of inhaled FITC-dextran out of the airspace was used to assess outside/in leak. Cytokines and neutrophils in bronchoalveolar lavage were assayed with ELISAs and cytospins respectively. Viral RNA level was assessed with RT-PCR and protein level assessed by ELISA. Main Results PKD inhibition prevented airway barrier dysfunction and pro-inflammatory cytokine release. Epithelial cells express PKD3, and PKD3 siRNA knock-down inhibited polyI:C induced cytokine production. Lung epithelial-specific deletion of PKD3 (CC10-Cre x PKD3-floxed mice) partially attenuated polyI:C-induced barrier disruption in vivo. Mechanistically, we found that PKD promoted cytokine mRNA transcription, not secretion, likely through activating the transcription factor Sp1. Finally, prophylactic CRT treatment of mice promoted barrier integrity during influenza virus infection and reduced viral burden. Conclusions Inhibiting PKD promotes barrier integrity, limit pathogenic cytokine levels, and restrict Influenza A Virus infection. Therefore, PKD is an attractive target for novel antiviral therapeutics.
Collapse
Affiliation(s)
- Janelle M Veazey
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, United States
| | - Sophia I Eliseeva
- Department of Medicine, Pulmonary and Critical Care, University of Rochester, Rochester, NY, United States
| | - Sara E Hillman
- Department of Medicine, Pulmonary and Critical Care, University of Rochester, Rochester, NY, United States
| | - Kristie Stiles
- Department of Medicine, Pulmonary and Critical Care, University of Rochester, Rochester, NY, United States
| | - Timothy R Smyth
- Department of Environmental Medicine, University of Rochester, Rochester, NY, United States
| | | | - Erika J Tillotson
- Department of Biology, Cornell University, Ithaca, NY, United States
| | - Dave J Topham
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, United States
| | - Timothy J Chapman
- Center for Infectious Disease and Immunology, Rochester Regional Health, Rochester, NY, United States
| | - Steve N Georas
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, United States.,Department of Medicine, Pulmonary and Critical Care, University of Rochester, Rochester, NY, United States.,Department of Environmental Medicine, University of Rochester, Rochester, NY, United States
| |
Collapse
|
15
|
Riquelme E, Zhang Y, Zhang L, Montiel M, Zoltan M, Dong W, Quesada P, Sahin I, Chandra V, San Lucas A, Scheet P, Xu H, Hanash SM, Feng L, Burks JK, Do KA, Peterson CB, Nejman D, Tzeng CWD, Kim MP, Sears CL, Ajami N, Petrosino J, Wood LD, Maitra A, Straussman R, Katz M, White JR, Jenq R, Wargo J, McAllister F. Tumor Microbiome Diversity and Composition Influence Pancreatic Cancer Outcomes. Cell 2020; 178:795-806.e12. [PMID: 31398337 DOI: 10.1016/j.cell.2019.07.008] [Citation(s) in RCA: 973] [Impact Index Per Article: 194.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 03/06/2019] [Accepted: 07/08/2019] [Indexed: 12/16/2022]
Abstract
Most patients diagnosed with resected pancreatic adenocarcinoma (PDAC) survive less than 5 years, but a minor subset survives longer. Here, we dissect the role of the tumor microbiota and the immune system in influencing long-term survival. Using 16S rRNA gene sequencing, we analyzed the tumor microbiome composition in PDAC patients with short-term survival (STS) and long-term survival (LTS). We found higher alpha-diversity in the tumor microbiome of LTS patients and identified an intra-tumoral microbiome signature (Pseudoxanthomonas-Streptomyces-Saccharopolyspora-Bacillus clausii) highly predictive of long-term survivorship in both discovery and validation cohorts. Through human-into-mice fecal microbiota transplantation (FMT) experiments from STS, LTS, or control donors, we were able to differentially modulate the tumor microbiome and affect tumor growth as well as tumor immune infiltration. Our study demonstrates that PDAC microbiome composition, which cross-talks to the gut microbiome, influences the host immune response and natural history of the disease.
Collapse
Affiliation(s)
- Erick Riquelme
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Center for Integrative Biology, Faculty of Science, Universidad Mayor, Santiago, Chile
| | - Yu Zhang
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Liangliang Zhang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Maria Montiel
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michelle Zoltan
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wenli Dong
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pompeyo Quesada
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ismet Sahin
- Department of Engineering, Texas Southern University, Houston, TX, USA
| | - Vidhi Chandra
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anthony San Lucas
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paul Scheet
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hanwen Xu
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Samir M Hanash
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; McCombs Institute for the Early Detection and Treatment of Cancer, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lei Feng
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jared K Burks
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kim-Anh Do
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christine B Peterson
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Deborah Nejman
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ching-Wei D Tzeng
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael P Kim
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cynthia L Sears
- Departments of Medicine, Oncology and Molecular Microbiology & Immunology, Johns Hopkins University School of Medicine and the Bloomberg School of Public Health, Baltimore, MD, USA
| | - Nadim Ajami
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Joseph Petrosino
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Laura D Wood
- Department of Pathology and The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anirban Maitra
- Sheikh Ahmed Pancreatic Cancer Research Center, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ravid Straussman
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Matthew Katz
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Robert Jenq
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer Wargo
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Florencia McAllister
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
16
|
Lérias JR, Paraschoudi G, de Sousa E, Martins J, Condeço C, Figueiredo N, Carvalho C, Dodoo E, Castillo-Martin M, Beltrán A, Ligeiro D, Rao M, Zumla A, Maeurer M. Microbes as Master Immunomodulators: Immunopathology, Cancer and Personalized Immunotherapies. Front Cell Dev Biol 2020; 7:362. [PMID: 32039196 PMCID: PMC6989410 DOI: 10.3389/fcell.2019.00362] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 12/12/2019] [Indexed: 12/12/2022] Open
Abstract
The intricate interplay between the immune system and microbes is an essential part of the physiological homeostasis in health and disease. Immunological recognition of commensal microbes, such as bacterial species resident in the gut or lung as well as dormant viral species, i.e., cytomegalovirus (CMV) or Epstein-Barr virus (EBV), in combination with a balanced immune regulation, is central to achieve immune-protection. Emerging evidence suggests that immune responses primed to guard against commensal microbes may cause unexpected pathological outcomes, e.g., chronic inflammation and/or malignant transformation. Furthermore, translocation of immune cells from one anatomical compartment to another, i.e., the gut-lung axis via the lymphatics or blood has been identified as an important factor in perpetrating systemic inflammation, tissue destruction, as well as modulating host-protective immune responses. We present in this review immune response patterns to pathogenic as well as non-pathogenic microbes and how these immune-recognition profiles affect local immune responses or malignant transformation. We discuss personalized immunological therapies which, directly or indirectly, target host biological pathways modulated by antimicrobial immune responses.
Collapse
Affiliation(s)
- Joana R. Lérias
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | | | - Eric de Sousa
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - João Martins
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Carolina Condeço
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Nuno Figueiredo
- Digestive Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Carlos Carvalho
- Digestive Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | | | | | - Antonio Beltrán
- Department of Pathology, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Dário Ligeiro
- Lisbon Centre for Blood and Transplantation, Instituto Português do Sangue e Transplantação, Lisbon, Portugal
| | - Martin Rao
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Alimuddin Zumla
- Division of Infection and Immunity, NIHR Biomedical Research Centre, University College London Hospitals NHS Foundation Trust, University College London, London, United Kingdom
| | - Markus Maeurer
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| |
Collapse
|
17
|
Yoon TW, Kim YI, Cho H, Brand DD, Rosloniec EF, Myers LK, Postlethwaite AE, Hasty KA, Stuart JM, Yi AK. Ameliorating effects of Gö6976, a pharmacological agent that inhibits protein kinase D, on collagen-induced arthritis. PLoS One 2019; 14:e0226145. [PMID: 31809526 PMCID: PMC6897462 DOI: 10.1371/journal.pone.0226145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 11/20/2019] [Indexed: 12/16/2022] Open
Abstract
Toll-like receptor (TLR) signaling can contribute to the pathogenesis of arthritis. Disruption of TLR signaling at early stages of arthritis might thereby provide an opportunity to halt the disease progression and ameliorate outcomes. We previously found that Gö6976 inhibits TLR-mediated cytokine production in human and mouse macrophages by inhibiting TLR-dependent activation of protein kinase D1 (PKD1), and that PKD1 is essential for proinflammatory responses mediated by MyD88-dependent TLRs. In this study, we investigated whether PKD1 contributes to TLR-mediated proinflammatory responses in human synovial cells, and whether Gö6976 treatment can suppress the development and progression of type II collagen (CII)-induced arthritis (CIA) in mouse. We found that TLR/IL-1R ligands induced activation of PKD1 in human fibroblast-like synoviocytes (HFLS). TLR/IL-1R-induced expression of cytokines/chemokines was substantially inhibited in Gö6976-treated HFLS and PKD1-knockdown HFLS. In addition, serum levels of anti-CII IgG antibodies, and the incidence and severity of arthritis after CII immunization were significantly reduced in mice treated daily with Gö6976. Synergistic effects of T-cell receptor and TLR, as well as TLR alone, on spleen cell proliferation and cytokine production were significantly inhibited in the presence of Gö6976. Our results suggest a possibility that ameliorating effects of Gö6976 on CIA may be due to its ability to inhibit TLR/IL-1R-activated PKD1, which might play an important role in proinflammatory responses in arthritis, and that PKD1 could be a therapeutic target for inflammatory arthritis.
Collapse
Affiliation(s)
- Tae Won Yoon
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Young-In Kim
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Hongsik Cho
- Department of Orthopedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - David D. Brand
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Veterans Affairs Medical Center-Memphis, Memphis, Tennessee, United States of America
| | - Edward F. Rosloniec
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Veterans Affairs Medical Center-Memphis, Memphis, Tennessee, United States of America
| | - Linda K. Myers
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Arnold E. Postlethwaite
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Veterans Affairs Medical Center-Memphis, Memphis, Tennessee, United States of America
| | - Karen A. Hasty
- Department of Orthopedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Veterans Affairs Medical Center-Memphis, Memphis, Tennessee, United States of America
| | - John M. Stuart
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Veterans Affairs Medical Center-Memphis, Memphis, Tennessee, United States of America
| | - Ae-Kyung Yi
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
18
|
Cho H, Bhatti FUR, Hasty KA, Yi AK. Nanosome-Mediated Delivery Of Protein Kinase D Inhibitor Protects Chondrocytes From Interleukin-1β-Induced Stress And Apoptotic Death. Int J Nanomedicine 2019; 14:8835-8846. [PMID: 31806974 PMCID: PMC6857658 DOI: 10.2147/ijn.s218901] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 08/30/2019] [Indexed: 01/10/2023] Open
Abstract
Background Inflammatory stress caused by protein kinase D (PKD) plays a critical role in damaging chondrocytes and extracellular matrix (ECM) during osteoarthritis (OA). The PKD inhibitor (PKDi) (CRT0066101) has been used to overcome inflammation in different cell types. However, the efficacy of a therapeutic drug can be limited due to off-target distribution, slow cellular internalization, and limited lysosomal escape. In order to overcome this issue, we developed nanosomes carrying CRT0066101 (PKDi-Nano) and tested their efficacy in vitro in chondrocytes. Methods Chondrocytes were subjected to IL-1β-induced inflammatory stress treated with either PKDi or PKDi-Nano. Effects of treatment were measured in terms of cytotoxicity, cellular morphology, viability, apoptosis, phosphorylation of protein kinase B (Akt), and anabolic/catabolic gene expression analyses related to cartilage tissue. Results and Discussion The effects of PKDi-Nano treatment were more pronounced as compared to PKDi treatment. Cytotoxicity and apoptosis were significantly reduced following PKDi-Nano treatment (P < 0.001). Cellular morphology was also restored to normal size and shape. The viability of chondrocytes was significantly enhanced in PKDi-Nano-treated cells (P < 0.001). The data indicated that PKDi-Nano acted independently of the Akt pathway. Gene expression analyses revealed significant increases in the expression levels of anabolic genes with concomitant decreases in the level of catabolic genes. Our results indicate that PKDi-Nano attenuated the effects of IL-1β via the nuclear factor kappa-light-chain enhancer of activated B cells (NF-κB) pathway. Conclusion Taken together, these results suggest that PKDi-Nano can be used as a successful strategy to reduce IL1β-induced inflammatory stress in chondrocytes.
Collapse
Affiliation(s)
- Hongsik Cho
- Department of Orthopaedic Surgery and Biomedical Engineering, The University of Tennessee Health Science Center, Memphis, TN, USA.,Department of Orthopaedic Surgery, Campbell Clinic, Memphis, TN, USA.,151 Research Service, Veterans Affairs Medical Center, Memphis, TN, USA
| | - Fazal-Ur-Rehman Bhatti
- Department of Orthopaedic Surgery and Biomedical Engineering, The University of Tennessee Health Science Center, Memphis, TN, USA.,151 Research Service, Veterans Affairs Medical Center, Memphis, TN, USA
| | - Karen A Hasty
- Department of Orthopaedic Surgery and Biomedical Engineering, The University of Tennessee Health Science Center, Memphis, TN, USA.,Department of Orthopaedic Surgery, Campbell Clinic, Memphis, TN, USA.,151 Research Service, Veterans Affairs Medical Center, Memphis, TN, USA
| | - Ae-Kyung Yi
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
19
|
Li J, Lv H, Che YQ. Upregulated microRNA-31 inhibits oxidative stress-induced neuronal injury through the JAK/STAT3 pathway by binding to PKD1 in mice with ischemic stroke. J Cell Physiol 2019; 235:2414-2428. [PMID: 31517390 DOI: 10.1002/jcp.29146] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 08/26/2019] [Indexed: 12/21/2022]
Abstract
Ischemic stroke (IS), which is characterized by high morbidity, disability, and mortality, is recognized as a major cerebrovascular disease. MicroRNA-31 (miR-31) was reported to participate in the progression of brain disease. The present study was conducted in order to investigate the effect of miR-31 on oxidative stress-induced neuronal injury in IS mice with the involvement of protein kinase D1 (PKD1) and the JAK/STAT3 pathway. C57BL/6J mice were used to establish the middle cerebral artery occlusion (MCAO) model. Astrocytes were transfected with miR-31 mimic, miR-31 inhibitor, si-PKD1, or JAK-STAT3 pathway inhibitor. Following the establishment of an oxygen-glucose deprivation (OGD) model, the astrocytes were cocultured with neuronal OGD. Lower miR-31, higher PKD1 expressions, and activated JAK/STAT3 pathway were found in both the MCAO and OGD models. miR-31 could negatively target PKD1. In an MCAO model, overexpressing miR-31 and silencing PKD1 reduced neuronal injury, cerebral infarct volume, neuron loss, and oxidative stress injury, inhibited the activation of JAK/STAT3 pathway and the expressions of PKD1, interleukin (IL)-1β, IL-6, tumor necrosis factor-α, malondialdehyde, 4-HNE, 8-HOdG, caspase-3, and Bax, but increased the superoxide dismutase content. In the OGD model, overexpression of miR-31 and silencing of PKD1 attenuated oxidative stress-induced neuronal injury, and diminished the lactate dehydrogenase leakage and reactive oxygen species level, accompanied by elevated neuronal viability. These results indicate that miR-31 alleviates inflammatory response as well as an oxidative stress-induced neuronal injury in IS mice by downregulating PKD1 and JAK/STAT3 pathway.
Collapse
Affiliation(s)
- Jie Li
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Hui Lv
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Yu-Qin Che
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
20
|
Youssef I, Ricort JM. Deciphering the Role of Protein Kinase D1 (PKD1) in Cellular Proliferation. Mol Cancer Res 2019; 17:1961-1974. [PMID: 31311827 DOI: 10.1158/1541-7786.mcr-19-0125] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 06/05/2019] [Accepted: 07/11/2019] [Indexed: 11/16/2022]
Abstract
Protein kinase D1 (PKD1) is a serine/threonine kinase that belongs to the calcium/calmodulin-dependent kinase family, and is involved in multiple mechanisms implicated in tumor progression such as cell motility, invasion, proliferation, protein transport, and apoptosis. While it is expressed in most tissues in the normal state, PKD1 expression may increase or decrease during tumorigenesis, and its role in proliferation is context-dependent and poorly understood. In this review, we present and discuss the current landscape of studies investigating the role of PKD1 in the proliferation of both cancerous and normal cells. Indeed, as a potential therapeutic target, deciphering whether PKD1 exerts a pro- or antiproliferative effect, and under what conditions, is of paramount importance.
Collapse
Affiliation(s)
- Ilige Youssef
- Centre National de la Recherche Scientifique, CNRS UMR_8113, Laboratoire de Biologie et Pharmacologie Appliquée, Cachan, France.,École Normale Supérieure Paris-Saclay, Université Paris-Saclay, Cachan, France
| | - Jean-Marc Ricort
- Centre National de la Recherche Scientifique, CNRS UMR_8113, Laboratoire de Biologie et Pharmacologie Appliquée, Cachan, France. .,École Normale Supérieure Paris-Saclay, Université Paris-Saclay, Cachan, France.,Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, Paris, France
| |
Collapse
|
21
|
Zhang J, Zhang Y, Wang J, Zhang S, Zhao Y, Ren H, Chu Y, Feng L, Wang C. Protein kinase D3 promotes gastric cancer development through p65/6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 activation of glycolysis. Exp Cell Res 2019; 380:188-197. [PMID: 31026442 DOI: 10.1016/j.yexcr.2019.04.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 04/17/2019] [Accepted: 04/18/2019] [Indexed: 11/16/2022]
Abstract
Although serine/threonine-protein kinases are found to participate in a wide range of cancer progression, the involvement of protein kinase D3 (PRKD3) in gastric cancer has not been explored. Here, we investigated the role of PRKD3 in gastric cancer (GC) and its potential mechanisms. PRKD3 was over-expressed in gastric cancer tissues and cells. In vitro, PRKD3 ectopic expression accelerated the proliferation and growth of GES-1, SGC7901 and MKN-28 cells. By contrast, PRKD3 knockdown suppressed the proliferation of SGC7901 and MKN-28 GC cells. In vivo, xenograted tumorigenesis was blunted by PRKD3 silencing. Mechanistically, PRKD3 up-regulated 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) and activated glycolysis as shown by increased glucose consumption and lactate production. Knockdown of PFKFB3 suppressed the glycolysis in gastric cancer cells with highly expressed PRKD3 but not in PRKD3 silenced cells. PRKD3 over-expression induced phosphorylation of p65 at serine 536 was critical for the up-regulation of glycolytic enzyme PFKFB3. Furthermore, PRKD and PFKFB3 inhibitor suppressed the viability of GC cells. Our results suggest that targeting PRKD3/p65/PFKFB3 cascade maybe a promising therapeutic strategy for gastric cancer.
Collapse
Affiliation(s)
- Jianwei Zhang
- Pancreatic and Gastric Surgery Department, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yajing Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jiaqi Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shuisheng Zhang
- Pancreatic and Gastric Surgery Department, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yajie Zhao
- Pancreatic and Gastric Surgery Department, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Hu Ren
- Pancreatic and Gastric Surgery Department, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yunmian Chu
- Pancreatic and Gastric Surgery Department, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lin Feng
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Chengfeng Wang
- Pancreatic and Gastric Surgery Department, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
22
|
Baker J, Falconer AMD, Wilkinson DJ, Europe-Finner GN, Litherland GJ, Rowan AD. Protein kinase D3 modulates MMP1 and MMP13 expression in human chondrocytes. PLoS One 2018; 13:e0195864. [PMID: 29652915 PMCID: PMC5898748 DOI: 10.1371/journal.pone.0195864] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 03/31/2018] [Indexed: 01/02/2023] Open
Abstract
Many catabolic stimuli, including interleukin-1 (IL-1) in combination with oncostatin M (OSM), promote cartilage breakdown via the induction of collagen-degrading collagenases such as matrix metalloproteinase 1 (MMP1) and MMP13 in human articular chondrocytes. Indeed, joint diseases with an inflammatory component are characterised by excessive extracellular matrix (ECM) catabolism. Importantly, protein kinase C (PKC) signalling has a primary role in cytokine-induced MMP1/13 expression, and is known to regulate cellular functions associated with pathologies involving ECM remodelling. At present, substrates downstream of PKC remain undefined. Herein, we show that both IL-1- and OSM-induced phosphorylation of protein kinase D (PKD) in human chondrocytes is strongly associated with signalling via the atypical PKCι isoform. Consequently, inhibiting PKD activation with a pan-PKD inhibitor significantly reduced the expression of MMP1/13. Specific gene silencing of the PKD isoforms revealed that only PKD3 (PRKD3) depletion mirrored the observed MMP repression, indicative of the pharmacological inhibitor specifically affecting only this isoform. PRKD3 silencing was also shown to reduce serine phosphorylation of signal transducer and activator of transcription 3 (STAT3) as well as phosphorylation of all three mitogen-activated protein kinase groups. This altered signalling following PRKD3 silencing led to a significant reduction in the expression of the activator protein-1 (AP-1) genes FOS and JUN, critical for the induction of many MMPs including MMP1/13. Furthermore, the AP-1 factor activating transcription factor 3 (ATF3) was also reduced concomitant with the observed reduction in MMP13 expression. Taken together, we highlight an important role for PKD3 in the pro-inflammatory signalling that promotes cartilage destruction.
Collapse
Affiliation(s)
- Jonathan Baker
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Adrian M. D. Falconer
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - David J. Wilkinson
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - G. Nicholas Europe-Finner
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Gary J. Litherland
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Andrew D. Rowan
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
- * E-mail:
| |
Collapse
|
23
|
Plastira I, Bernhart E, Goeritzer M, DeVaney T, Reicher H, Hammer A, Lohberger B, Wintersperger A, Zucol B, Graier WF, Kratky D, Malle E, Sattler W. Lysophosphatidic acid via LPA-receptor 5/protein kinase D-dependent pathways induces a motile and pro-inflammatory microglial phenotype. J Neuroinflammation 2017; 14:253. [PMID: 29258556 PMCID: PMC5735906 DOI: 10.1186/s12974-017-1024-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 12/06/2017] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Extracellular lysophosphatidic acid (LPA) species transmit signals via six different G protein-coupled receptors (LPAR1-6) and are indispensible for brain development and function of the nervous system. However, under neuroinflammatory conditions or brain damage, LPA levels increase, thereby inducing signaling cascades that counteract brain function. We describe a critical role for 1-oleyl-2-hydroxy-sn-glycero-3-phosphate (termed "LPA" throughout our study) in mediating a motile and pro-inflammatory microglial phenotype via LPAR5 that couples to protein kinase D (PKD)-mediated pathways. METHODS Using the xCELLigence system and time-lapse microscopy, we investigated the migrational response of microglial cells. Different M1 and M2 markers were analyzed by confocal microscopy, flow cytometry, and immunoblotting. Using qPCR and ELISA, we studied the expression of migratory genes and quantitated the secretion of pro-inflammatory cytokines and chemokines, respectively. Different transcription factors that promote the regulation of pro-inflammatory genes were analyzed by western blot. Reactive oxygen species (ROS) and nitric oxide (NO) production, phagocytosis, and microglial cytotoxicity were determined using commercially available assay kits. RESULTS LPA induces MAPK family and AKT activation and pro-inflammatory transcription factors' phosphorylation (NF-κB, c-Jun, STAT1, and STAT3) that were inhibited by both LPAR5 and PKD family antagonists. LPA increases migratory capacity, induces secretion of pro-inflammatory cytokines and chemokines and expression of M1 markers, enhances production of ROS and NO by microglia, and augments cytotoxicity of microglial cell-conditioned medium towards neurons. The PKD family inhibitor blunted all of these effects. We propose that interference with this signaling axis could aid in the development of new therapeutic approaches to control neuroinflammation under conditions of overshooting LPA production. CONCLUSIONS In the present study, we show that inflammatory LPA levels increased the migratory response of microglia and promoted a pro-inflammatory phenotype via the LPAR5/PKD axis. Interference with this signaling axis reduced microglial migration, blunted microglial cytotoxicity, and abrogated the expression and secretion of pro-inflammatory mediators.
Collapse
Affiliation(s)
- I. Plastira
- 0000 0000 8988 2476grid.11598.34Institute of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria
| | - E. Bernhart
- 0000 0000 8988 2476grid.11598.34Institute of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria
| | - M. Goeritzer
- 0000 0000 8988 2476grid.11598.34Institute of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria ,grid.452216.6BioTechMed-Graz, Graz, Austria
| | - T. DeVaney
- 0000 0000 8988 2476grid.11598.34Institute of Biophysics, Medical University of Graz, Graz, Austria
| | - H. Reicher
- 0000 0000 8988 2476grid.11598.34Institute of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria
| | - A. Hammer
- 0000 0000 8988 2476grid.11598.34Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - B. Lohberger
- 0000 0000 8988 2476grid.11598.34Department of Orthopedic Surgery, Medical University of Graz, Graz, Austria
| | - A. Wintersperger
- 0000 0000 8988 2476grid.11598.34Institute of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria
| | - B. Zucol
- 0000 0000 8988 2476grid.11598.34Institute of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria
| | - W. F. Graier
- 0000 0000 8988 2476grid.11598.34Institute of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria ,grid.452216.6BioTechMed-Graz, Graz, Austria
| | - D. Kratky
- 0000 0000 8988 2476grid.11598.34Institute of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria ,grid.452216.6BioTechMed-Graz, Graz, Austria
| | - E. Malle
- 0000 0000 8988 2476grid.11598.34Institute of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria
| | - W. Sattler
- 0000 0000 8988 2476grid.11598.34Institute of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria ,grid.452216.6BioTechMed-Graz, Graz, Austria
| |
Collapse
|
24
|
Upadhyay K, Park JE, Yoon TW, Halder P, Kim YI, Metcalfe V, Talati AJ, English BK, Yi AK. Group B Streptococci Induce Proinflammatory Responses via a Protein Kinase D1-Dependent Pathway. THE JOURNAL OF IMMUNOLOGY 2017; 198:4448-4457. [PMID: 28461572 DOI: 10.4049/jimmunol.1601089] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 04/02/2017] [Indexed: 12/31/2022]
Abstract
Group B streptococci (GBS) are one of the leading causes of life-threatening illness in neonates. Proinflammatory responses to GBS mediated through host innate immune receptors play a critical role in the disease manifestation. However, the mechanisms involved in proinflammatory responses against GBS, as well as the contribution of signaling modulators involved in host immune defense, have not been fully elucidated. In the present study, we investigated the role of protein kinase D (PKD)1 in the proinflammatory responses to GBS. We found that both live and antibiotic-killed GBS induce activation of PKD1 through a pathway that is dependent on the TLR signaling adaptor MyD88 and its downstream kinase IL-1R-associated kinase 1, but independent of TNFR-associated factor 6. Our studies using pharmacological PKD inhibitors and PKD1-knockdown macrophages revealed that PKD1 is indispensable for GBS-mediated activation of MAPKs and NF-κB and subsequent expression of proinflammatory mediators. Furthermore, systemic administration of a PKD inhibitor protects d-galactosamine-sensitized mice from shock-mediated death caused by antibiotic-killed GBS. These findings imply that PKD1 plays a critical regulatory role in GBS-induced proinflammatory reactions and sepsis, and inhibition of PKD1 activation together with antibiotic treatment in GBS-infected neonates could be an effective way to control GBS diseases.
Collapse
Affiliation(s)
- Kirtikumar Upadhyay
- Department of Pediatrics, The University of Tennessee Health Science Center, Memphis, TN 38163.,Department of Obstetrics and Gynecology, The University of Tennessee Health Science Center, Memphis, TN 38163.,Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, TN 38103
| | - Jeoung-Eun Park
- Department of Pediatrics, The University of Tennessee Health Science Center, Memphis, TN 38163
| | - Tae Won Yoon
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN 38163; and
| | - Priyanka Halder
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN 38163; and
| | - Young-In Kim
- Department of Pediatrics, The University of Tennessee Health Science Center, Memphis, TN 38163.,Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, TN 38103
| | - Victoria Metcalfe
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN 38163; and
| | - Ajay J Talati
- Department of Pediatrics, The University of Tennessee Health Science Center, Memphis, TN 38163.,Department of Obstetrics and Gynecology, The University of Tennessee Health Science Center, Memphis, TN 38163.,Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, TN 38103
| | - B Keith English
- Department of Pediatrics and Human Development, Michigan State University, Lansing, MI 48912
| | - Ae-Kyung Yi
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN 38163; and
| |
Collapse
|
25
|
Protein Kinase D2 Protects against Acute Colitis Induced by Dextran Sulfate Sodium in Mice. Sci Rep 2016; 6:34079. [PMID: 27659202 PMCID: PMC5034322 DOI: 10.1038/srep34079] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 09/07/2016] [Indexed: 12/11/2022] Open
Abstract
Inflammatory bowel disease is characterized by dysregulation of the mucosal immune system resulting from impaired intestinal epithelial barrier function. Protein kinase D2 has been implicated in the regulation of immune responses. The present study was to define PKD2 might affect murine colitis. Colitis was induced in wild-type mice (PKD2WT/WT) and PKD2 catalytic activity deficient mice (PKD2SSAA/SSAA) with dextran sulfate sodium. PKD2SSAA-knockin mice displayed catalytic activity deficiency and increased susceptibility to DSS-induced colitis with enhanced weight loss, colonic inflammation compared with PKD2WT/WT mice. Furthermore, crucial inflammatory cytokines mRNA levels in PKD2SSAA-knockin mice were higher than controls accompanied with down-regulation of ZO-1, MUC2 and intestinal barrier dysfunction. However, there were no differences in the proliferation or apoptosis of intestinal epithelial cells in PKD2SSAA-knockin mice compared with wild-type controls. In addition, PKD2 expression was repressed in patients with IBD compared with healthy controls. These studies suggested that activation of PKD2 in the colonic epithelium microenvironment may contribute to protect against DSS-induced colitis through regulation of intestinal mucosal immunity and barrier function.
Collapse
|
26
|
Najibi M, Labed SA, Visvikis O, Irazoqui JE. An Evolutionarily Conserved PLC-PKD-TFEB Pathway for Host Defense. Cell Rep 2016; 15:1728-42. [PMID: 27184844 DOI: 10.1016/j.celrep.2016.04.052] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 01/28/2016] [Accepted: 04/13/2016] [Indexed: 12/18/2022] Open
Abstract
The mechanisms that tightly control the transcription of host defense genes have not been fully elucidated. We previously identified TFEB as a transcription factor important for host defense, but the mechanisms that regulate TFEB during infection remained unknown. Here, we used C. elegans to discover a pathway that activates TFEB during infection. Gene dkf-1, which encodes a homolog of protein kinase D (PKD), was required for TFEB activation in nematodes infected with Staphylococcus aureus. Conversely, pharmacological activation of PKD was sufficient to activate TFEB. Furthermore, phospholipase C (PLC) gene plc-1 was also required for TFEB activation, downstream of Gαq homolog egl-30 and upstream of dkf-1. Using reverse and chemical genetics, we discovered a similar PLC-PKD-TFEB axis in Salmonella-infected mouse macrophages. In addition, PKCα was required in macrophages. These observations reveal a previously unknown host defense signaling pathway, which has been conserved across one billion years of evolution.
Collapse
Affiliation(s)
- Mehran Najibi
- Laboratory of Comparative Immunology, Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, MA 02114, USA
| | - Sid Ahmed Labed
- Laboratory of Comparative Immunology, Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, MA 02114, USA
| | - Orane Visvikis
- Laboratory of Comparative Immunology, Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, MA 02114, USA
| | - Javier Elbio Irazoqui
- Laboratory of Comparative Immunology, Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
27
|
Varga A, Gyulavári P, Greff Z, Futosi K, Németh T, Simon-Szabó L, Kerekes K, Szántai-Kis C, Brauswetter D, Kokas M, Borbély G, Erdei A, Mócsai A, Kéri G, Vántus T. Targeting vascular endothelial growth factor receptor 2 and protein kinase D1 related pathways by a multiple kinase inhibitor in angiogenesis and inflammation related processes in vitro. PLoS One 2015; 10:e0124234. [PMID: 25874616 PMCID: PMC4396990 DOI: 10.1371/journal.pone.0124234] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 02/27/2015] [Indexed: 12/18/2022] Open
Abstract
Emerging evidence suggests that the vascular endothelial growth factor receptor 2 (VEGFR2) and protein kinase D1 (PKD1) signaling axis plays a critical role in normal and pathological angiogenesis and inflammation related processes. Despite all efforts, the currently available therapeutic interventions are limited. Prior studies have also proved that a multiple target inhibitor can be more efficient compared to a single target one. Therefore, development of novel inflammatory pathway-specific inhibitors would be of great value. To test this possibility, we screened our molecular library using recombinant kinase assays and identified the previously described compound VCC251801 with strong inhibitory effect on both VEGFR2 and PKD1. We further analyzed the effect of VCC251801 in the endothelium-derived EA.hy926 cell line and in different inflammatory cell types. In EA.hy926 cells, VCC251801 potently inhibited the intracellular activation and signaling of VEGFR2 and PKD1 which inhibition eventually resulted in diminished cell proliferation. In this model, our compound was also an efficient inhibitor of in vitro angiogenesis by interfering with endothelial cell migration and tube formation processes. Our results from functional assays in inflammatory cellular models such as neutrophils and mast cells suggested an anti-inflammatory effect of VCC251801. The neutrophil study showed that VCC251801 specifically blocked the immobilized immune-complex and the adhesion dependent TNF-α -fibrinogen stimulated neutrophil activation. Furthermore, similar results were found in mast cell degranulation assay where VCC251801 caused significant reduction of mast cell response. In summary, we described a novel function of a multiple kinase inhibitor which strongly inhibits the VEGFR2-PKD1 signaling and might be a novel inhibitor of pathological inflammatory pathways.
Collapse
Affiliation(s)
- Attila Varga
- Pathobiochemistry Research Group, Hungarian Academy of Sciences—Semmelweis University, Budapest, Hungary
| | - Pál Gyulavári
- Pathobiochemistry Research Group, Hungarian Academy of Sciences—Semmelweis University, Budapest, Hungary
| | | | - Krisztina Futosi
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Tamás Németh
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Laura Simon-Szabó
- Department of Immunology, Eötvös Loránd University, Budapest, Hungary
| | - Krisztina Kerekes
- Department of Immunology, Eötvös Loránd University, Budapest, Hungary
| | | | - Diána Brauswetter
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Márton Kokas
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Gábor Borbély
- Pathobiochemistry Research Group, Hungarian Academy of Sciences—Semmelweis University, Budapest, Hungary
| | - Anna Erdei
- Department of Immunology, Eötvös Loránd University, Budapest, Hungary
| | - Attila Mócsai
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - György Kéri
- Pathobiochemistry Research Group, Hungarian Academy of Sciences—Semmelweis University, Budapest, Hungary
- Vichem Chemie Research Ltd., Budapest, Hungary
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Tibor Vántus
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
- * E-mail:
| |
Collapse
|
28
|
Xu W, Wang Y, Li S, Ke Z, Yan Y, Li S, Xing Z, Wang C, Zeng F, Liu R, Deng F. Efficient gene and siRNA delivery with cationic polyphosphoramide with amino moieties in the main chain. RSC Adv 2015. [DOI: 10.1039/c5ra02721g] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
A novel cation polyphosphoramide with amino moieties in the main chain was synthesized, which can be used as efficient carriers for plasmid and siRNA.
Collapse
|
29
|
Protein kinase D is increased and activated in lung epithelial cells and macrophages in idiopathic pulmonary fibrosis. PLoS One 2014; 9:e101983. [PMID: 25000413 PMCID: PMC4084945 DOI: 10.1371/journal.pone.0101983] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 06/12/2014] [Indexed: 01/13/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a relentlessly progressive and usually fatal lung disease of unknown etiology for which no effective treatments currently exist. Hence, there is a profound need for the identification of novel drugable targets to develop more specific and efficacious therapeutic intervention in IPF. In this study, we performed immunohistochemical analyses to assess the cell type-specific expression and activation of protein kinase D (PKD) family kinases in normal and IPF lung tissue sections. We also analyzed PKD activation and function in human lung epithelial cells. We found that PKD family kinases (PKD1, PKD2 and PKD3) were increased and activated in the hyperplastic and regenerative alveolar epithelial cells lining remodeled fibrotic alveolar septa and/or fibroblast foci in IPF lungs compared with normal controls. We also found that PKD family kinases were increased and activated in alveolar macrophages, bronchiolar epithelium, and honeycomb cysts in IPF lungs. Interestingly, PKD1 was highly expressed and activated in the cilia of IPF bronchiolar epithelial cells, while PKD2 and PKD3 were expressed in the cell cytoplasm and nuclei. In contrast, PKD family kinases were not apparently increased and activated in IPF fibroblasts or myofibroblasts. We lastly found that PKD was predominantly activated by poly-L-arginine, lysophosphatidic acid and thrombin in human lung epithelial cells and that PKD promoted epithelial barrier dysfunction. These findings suggest that PKD may participate in the pathogenesis of IPF and may be a novel target for therapeutic intervention in this disease.
Collapse
|
30
|
Rezaee F, DeSando SA, Ivanov AI, Chapman TJ, Knowlden SA, Beck LA, Georas SN. Sustained protein kinase D activation mediates respiratory syncytial virus-induced airway barrier disruption. J Virol 2013; 87:11088-95. [PMID: 23926335 PMCID: PMC3807305 DOI: 10.1128/jvi.01573-13] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 07/29/2013] [Indexed: 01/08/2023] Open
Abstract
Understanding the regulation of airway epithelial barrier function is a new frontier in asthma and respiratory viral infections. Despite recent progress, little is known about how respiratory syncytial virus (RSV) acts at mucosal sites, and very little is known about its ability to influence airway epithelial barrier function. Here, we studied the effect of RSV infection on the airway epithelial barrier using model epithelia. 16HBE14o- bronchial epithelial cells were grown on Transwell inserts and infected with RSV strain A2. We analyzed (i) epithelial apical junction complex (AJC) function, measuring transepithelial electrical resistance (TEER) and permeability to fluorescein isothiocyanate (FITC)-conjugated dextran, and (ii) AJC structure using immunofluorescent staining. Cells were pretreated or not with protein kinase D (PKD) inhibitors. UV-irradiated RSV served as a negative control. RSV infection led to a significant reduction in TEER and increase in permeability. Additionally it caused disruption of the AJC and remodeling of the apical actin cytoskeleton. Pretreatment with two structurally unrelated PKD inhibitors markedly attenuated RSV-induced effects. RSV induced phosphorylation of the actin binding protein cortactin in a PKD-dependent manner. UV-inactivated RSV had no effect on AJC function or structure. Our results suggest that RSV-induced airway epithelial barrier disruption involves PKD-dependent actin cytoskeletal remodeling, possibly dependent on cortactin activation. Defining the mechanisms by which RSV disrupts epithelial structure and function should enhance our understanding of the association between respiratory viral infections, airway inflammation, and allergen sensitization. Impaired barrier function may open a potential new therapeutic target for RSV-mediated lung diseases.
Collapse
Affiliation(s)
- Fariba Rezaee
- Division of Pediatric Pulmonary Medicine, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| | - Samantha A. DeSando
- Division of Pediatric Pulmonary Medicine, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| | - Andrei I. Ivanov
- Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Timothy J. Chapman
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | - Sara A. Knowlden
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | - Lisa A. Beck
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA
| | - Steve N. Georas
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| |
Collapse
|
31
|
Lacasse Y, Girard M, Cormier Y. Recent advances in hypersensitivity pneumonitis. Chest 2012; 142:208-217. [PMID: 22796841 DOI: 10.1378/chest.11-2479] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Hypersensitivity pneumonitis (HP) is a pulmonary disease with symptoms of dyspnea and cough resulting from the inhalation of an allergen to which the subject has been previously sensitized. The diagnosis of HP most often relies on an array of nonspecific clinical symptoms and signs developed in an appropriate setting, with the demonstration of interstitial markings on chest radiographs, serum precipitating antibodies against offending antigens, a lymphocytic alveolitis on BAL, and/or a granulomatous reaction on lung biopsies. The current classification of HP in acute, subacute, and chronic phases is now challenged, and a set of clinical predictors has been proposed. Nonspecific interstitial pneumonitis, usual interstitial pneumonia, and bronchiolitis obliterans organizing pneumonia may be the sole histologic expression of the disease. Presumably, like in idiopathic interstitial pneumonia, acute exacerbations of chronic HP may occur without further exposure to the offending antigen. New offending antigens, such as mycobacteria causing hot tub lung and metalworking fluid HP, have recently been identified and have stimulated further research in HP.
Collapse
Affiliation(s)
- Yves Lacasse
- Centre de Recherche, Institut Universitaire de Cardiologie et de Pneumologie de Québec (Hôpital Laval), Québec, QC, Canada.
| | - Mélissa Girard
- Centre de Recherche, Institut Universitaire de Cardiologie et de Pneumologie de Québec (Hôpital Laval), Québec, QC, Canada
| | - Yvon Cormier
- Centre de Recherche, Institut Universitaire de Cardiologie et de Pneumologie de Québec (Hôpital Laval), Québec, QC, Canada
| |
Collapse
|
32
|
A targeted library screen reveals a new inhibitor scaffold for protein kinase D. PLoS One 2012; 7:e44653. [PMID: 23028574 PMCID: PMC3445516 DOI: 10.1371/journal.pone.0044653] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 08/06/2012] [Indexed: 12/31/2022] Open
Abstract
Protein kinase D (PKD) has emerged as a potential therapeutic target in multiple pathological conditions, including cancer and heart diseases. Potent and selective small molecule inhibitors of PKD are valuable for dissecting PKD-mediated cellular signaling pathways and for therapeutic application. In this study, we evaluated a targeted library of 235 small organic kinase inhibitors for PKD1 inhibitory activity at a single concentration. Twenty-eight PKD inhibitory chemotypes were identified and six exhibited excellent PKD1 selectivity. Five of the six lead structures share a common scaffold, with compound 139 being the most potent and selective for PKD vs PKC and CAMK. Compound 139 was an ATP-competitive PKD1 inhibitor with a low double-digit nanomolar potency and was also cell-active. Kinase profiling analysis identified this class of small molecules as pan-PKD inhibitors, confirmed their selectivity again PKC and CAMK, and demonstrated an overall favorable selectivity profile that could be further enhanced through structural modification. Furthermore, using a PKD homology model based on similar protein kinase structures, docking modes for compound 139 were explored and compared to literature examples of PKD inhibition. Modeling of these compounds at the ATP-binding site of PKD was used to rationalize its high potency and provide the foundation for future further optimization. Accordingly, using biochemical screening of a small number of privileged scaffolds and computational modeling, we have identified a new core structure for highly potent PKD inhibition with promising selectivity against closely related kinases. These lead structures represent an excellent starting point for the further optimization and the design of selective and therapeutically effective small molecule inhibitors of PKD.
Collapse
|
33
|
Zou Z, Zeng F, Xu W, Wang C, Ke Z, Wang QJ, Deng F. PKD2 and PKD3 promote prostate cancer cell invasion by modulating NF-κB- and HDAC1-mediated expression and activation of uPA. J Cell Sci 2012; 125:4800-11. [PMID: 22797919 DOI: 10.1242/jcs.106542] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Although protein kinase D3 (PKD3) has been shown to contribute to prostate cancer cell growth and survival, the role of PKD in prostate cancer cell motility remains unclear. Here, we show that PKD2 and PKD3 promote nuclear factor kappa B (NF-κB) signaling and urokinase-type plasminogen activator (uPA) expression/activation, which are crucial for prostate cancer cell invasion. Silencing of endogenous PKD2 and/or PKD3 markedly decreased prostate cancer cell migration and invasion, reduced uPA and uPA receptor (uPAR) expression and increased plasminogen activator inhibitor-2 (PAI-2) expression. These results were further substantiated by the finding that PKD2 and PKD3 promoted the activity of uPA and matrix metalloproteinase 9 (MMP9). Furthermore, depletion of PKD2 and/or PKD3 decreased the level of binding of the p65 subunit of NF-κB to the promoter of the gene encoding uPA (PLAU), suppressing transcriptional activation of uPA. Endogenous PKD2 and PKD3 interacted with inhibitor of NF-κB (IκB) kinase β (IKKβ); PKD2 mainly regulated the phosphorylated IKK (pIKK)-phosphorylated IκB (pIκB)-IκB degradation cascade, p65 nuclear translocation, and phosphorylation of Ser276 on p65, whereas PKD3 was responsible for the phosphorylation of Ser536 on p65. Conversely, inhibition of uPA transactivation by PKD3 silencing was rescued by constitutive Ser536 p65 phosphorylation, and reduced tumor cell invasion resulting from PKD2 or PKD3 silencing was rescued by ectopic expression of p65. Interestingly, PKD3 interacted with histone deacetylase 1 (HDAC1), suppressing HDAC1 expression and decreasing its binding to the uPA promoter. Moreover, depletion of HDAC1 resulted in recovery of uPA transactivation in PKD3-knockdown cells. Taken together, these data suggest that PKD2 and PKD3 coordinate to promote prostate cancer cell invasion through p65 NF-κB- and HDAC1-mediated expression and activation of uPA.
Collapse
Affiliation(s)
- Zhipeng Zou
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | | | | | | | | | | | | |
Collapse
|
34
|
Rezaee F, Meednu N, Emo JA, Saatian B, Chapman TJ, Naydenov NG, De Benedetto A, Beck LA, Ivanov AI, Georas SN. Polyinosinic:polycytidylic acid induces protein kinase D-dependent disassembly of apical junctions and barrier dysfunction in airway epithelial cells. J Allergy Clin Immunol 2011; 128:1216-1224.e11. [PMID: 21996340 DOI: 10.1016/j.jaci.2011.08.035] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Revised: 08/25/2011] [Accepted: 08/29/2011] [Indexed: 12/21/2022]
Abstract
BACKGROUND Disruption of the epithelial barrier might be a risk factor for allergen sensitization and asthma. Viral respiratory tract infections are strongly associated with asthma exacerbation, but the effects of respiratory viruses on airway epithelial barrier function are not well understood. Many viruses generate double-stranded RNA, which can lead to airway inflammation and initiate an antiviral immune response. OBJECTIVES We investigated the effects of the synthetic double-stranded RNA polyinosinic:polycytidylic acid (polyI:C) on the structure and function of the airway epithelial barrier in vitro. METHODS 16HBE14o- human bronchial epithelial cells and primary airway epithelial cells at an air-liquid interface were grown to confluence on Transwell inserts and exposed to polyI:C. We studied epithelial barrier function by measuring transepithelial electrical resistance and paracellular flux of fluorescent markers and structure of epithelial apical junctions by means of immunofluorescence microscopy. RESULTS PolyI:C induced a profound decrease in transepithelial electrical resistance and increase in paracellular permeability. Immunofluorescence microscopy revealed markedly reduced junctional localization of zonula occludens-1, occludin, E-cadherin, β-catenin, and disorganization of junction-associated actin filaments. PolyI:C induced protein kinase D (PKD) phosphorylation, and a PKD antagonist attenuated polyI:C-induced disassembly of apical junctions and barrier dysfunction. CONCLUSIONS PolyI:C has a powerful and previously unsuspected disruptive effect on the airway epithelial barrier. PolyI:C-dependent barrier disruption is mediated by disassembly of epithelial apical junctions, which is dependent on PKD signaling. These findings suggest a new mechanism potentially underlying the associations between viral respiratory tract infections, airway inflammation, and allergen sensitization.
Collapse
Affiliation(s)
- Fariba Rezaee
- Division of Pediatric Pulmonary, Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14610, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Rozengurt E. Protein kinase D signaling: multiple biological functions in health and disease. Physiology (Bethesda) 2011; 26:23-33. [PMID: 21357900 DOI: 10.1152/physiol.00037.2010] [Citation(s) in RCA: 184] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Protein kinase D (PKD) is an evolutionarily conserved protein kinase family with structural, enzymological, and regulatory properties different from the PKC family members. Signaling through PKD is induced by a remarkable number of stimuli, including G-protein-coupled receptor agonists and polypeptide growth factors. PKD1, the most studied member of the family, is increasingly implicated in the regulation of a complex array of fundamental biological processes, including signal transduction, cell proliferation and differentiation, membrane trafficking, secretion, immune regulation, cardiac hypertrophy and contraction, angiogenesis, and cancer. PKD mediates such a diverse array of normal and abnormal biological functions via dynamic changes in its spatial and temporal localization, combined with its distinct substrate specificity. Studies on PKD thus far indicate a striking diversity of both its signal generation and distribution and its potential for complex regulatory interactions with multiple downstream pathways, often regulating the subcellular localization of its targets.
Collapse
Affiliation(s)
- Enrique Rozengurt
- Department of Medicine, Division of Digestive Diseases, David Geffen School of Medicine, CURE: Digestive Diseases Research Center and Molecular Biology Institute, University of California, Los Angeles, California, USA.
| |
Collapse
|