1
|
Umumararungu T, Nyandwi JB, Katandula J, Twizeyimana E, Claude Tomani J, Gahamanyi N, Ishimwe N, Olawode EO, Habarurema G, Mpenda M, Uyisenga JP, Saeed SI. Current status of the small molecule anti-HIV drugs in the pipeline or recently approved. Bioorg Med Chem 2024; 111:117860. [PMID: 39094527 DOI: 10.1016/j.bmc.2024.117860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/04/2024]
Abstract
Human Immunodeficiency Virus (HIV) is the causative agent of Acquired Immunodeficiency Syndrome (AIDS) with high morbidity and mortality rates. Treatment of AIDS/HIV is being complicated by increasing resistance to currently used antiretroviral (ARV) drugs, mainly in low- and middle-income countries (LMICs) due to drug misuse, poor drug supply and poor treatment monitoring. However, progress has been made in the development of new ARV drugs, targeting different HIV components (Fig. 1). This review aims at presenting and discussing the progress made towards the discovery of new ARVs that are at different stages of clinical trials as of July 2024. For each compound, the mechanism of action, target biomolecule, genes associated with resistance, efficacy and safety, class, and phase of clinical trial are discussed. These compounds include analogues of nucleoside reverse transcriptase inhibitors (NRTIs) - islatravir and censavudine; non-nucleoside reverse transcriptase inhibitors (NNRTIs) - Rilpivirine, elsulfavirine and doravirine; integrase inhibitors namely cabotegravir and dolutegravir and chemokine coreceptors 5 and 2 (CC5/CCR2) antagonists for example cenicriviroc. Also, fostemsavir is being developed as an attachment inhibitor while lenacapavir, VH4004280 and VH4011499 are capsid inhibitors. Others are maturation inhibitors such as GSK-254, GSK3532795, GSK3739937, GSK2838232, and other compounds labelled as miscellaneous (do not belong to the classical groups of anti-HIV drugs or to the newer classes) such as obefazimod and BIT225. There is a considerable progress in the development of new anti-HIV drugs and the effort will continue since HIV infections has no cure or vaccine till now. Efforts are needed to reduce the toxicity of available drugs or discover new drugs with new classes which can delay the development of resistance.
Collapse
Affiliation(s)
- Théoneste Umumararungu
- Department of Industrial Pharmacy, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Rwanda.
| | - Jean Baptiste Nyandwi
- Department of Pharmacology and Toxicology, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Rwanda; East African Community Regional Centre of Excellence for Vaccines, Immunization and Health Supply Chain Management, Kigali, Rwanda
| | - Jonathan Katandula
- Department of Pharmacology and Toxicology, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Rwanda
| | - Eric Twizeyimana
- Department of Physiology, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Rwanda
| | - Jean Claude Tomani
- Department of Chemistry, School of Science, College of Science and Technology, University of Rwanda, Rwanda
| | - Noël Gahamanyi
- Department of Biology, School of Science, College of Science and Technology, University of Rwanda, Rwanda
| | - Nestor Ishimwe
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Emmanuel Oladayo Olawode
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, 18301 N Miami Ave #1, Miami, FL 33169, USA
| | - Gratien Habarurema
- Department of Chemistry, School of Science, College of Science and Technology, University of Rwanda, Rwanda
| | - Matabishi Mpenda
- Department of Pharmacology and Toxicology, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Rwanda
| | - Jeanne Primitive Uyisenga
- Department of Biology, School of Science, College of Science and Technology, University of Rwanda, Rwanda
| | - Shamsaldeen Ibrahim Saeed
- Faculty of Veterinary Science, University of Nyala, P.O. Box: 155, Nyala, Sudan; Nanotechnology in Veterinary Medicine (NanoVet) Research Group, Faculty of Veterinary Medicine, University Malaysia Kelantan, Kelantan 16100, Pengkalan Chepa, Malaysia
| |
Collapse
|
2
|
Prins CA, de Oliveira FL, de Mello Coelho V, Dos Santos Ribeiro EB, de Almeida JS, Silva NMB, Almeida FM, Martinez AMB. Galectin-3 absence alters lymphocytes populations dynamics behavior and promotes functional recovery after spinal cord injury in mice. Exp Neurol 2024; 377:114785. [PMID: 38670250 DOI: 10.1016/j.expneurol.2024.114785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 04/02/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024]
Abstract
Spinal cord injury (SCI) results from various mechanisms that damage the nervous tissue and the blood-brain barrier, leading to sensory and motor function loss below the injury site. Unfortunately, current therapeutic approaches for SCI have limited efficacy in improving patients outcomes. Galectin-3, a protein whose expression increases after SCI, influences the neuroinflammatory response by favoring pro-inflammatory M1 macrophages and microglia, while inhibiting pro-regenerative M2 macrophages and microglia, which are crucial for inflammation resolution and tissue regeneration. Previous studies with Galectin-3 knock-out mice demonstrated enhanced motor recovery after SCI. The M1/M2 balance is strongly influenced by the predominant lymphocytic profiles (Th1, Th2, T Reg, Th17) and cytokines and chemokines released at the lesion site. The present study aimed to investigate how the absence of galectin-3 impacts the adaptive immune system cell population dynamics in various lymphoid spaces following a low thoracic spinal cord compression injury (T9-T10) using a 30 g vascular clip for one minute. It also aimed to assess its influence on the functional outcome in wild-type (WT)and Galectin-3 knock-out (GALNEG) mice. Histological analysis with hematoxylin-eosin and Luxol Fast Blue staining revealed that WT and GALNEG animals exhibit similar spinal cord morphology. The absence of galectin-3 does not affect the common neuroanatomy shared between the groups prompting us to analyze outcomes between both groups. Following our crush model, both groups lost motor and sensory functions below the lesion level. During a 42-day period, GALNEG mice demonstrated superior locomotor recovery in the Basso Mouse Scale (BMS) gait analysis and enhanced motor coordination performance in the ladder rung walk test (LRW) compared to WT mice. GALNEG mice also exhibited better sensory recovery, and their electrophysiological parameters suggested a higher number of functional axons with faster nerve conduction. Seven days after injury, flow cytometry of thymus, spleen, and blood revealed an increased number of T Reg and Th2 cells, accompanied by a decrease in Th1 and Th17 cells in GALNEG mice. Immunohistochemistry conducted on the same day exhibited an increased number of Th2 and T Reg cells around the GALNEG's spinal cord lesion site. At 42-day dpi immunohistochemistry analyses displayed reduced astrogliosis and greater axon preservation in GALNEG's spinal cord seem as a reduction of GFAP immunostaining and an increase in NFH immunostaining, respectively. In conclusion, GALNEG mice exhibited better functional recovery attributed to the milder pro-inflammatory influence, compensated by a higher quantity of T Reg and Th2 cells. These findings suggest that galectin-3 plays a crucial role in the immune response after spinal cord injury and could be a potential target for clinical therapeutic interventions.
Collapse
Affiliation(s)
- Caio Andrade Prins
- Laboratório de Neurodegeneração e Reparo, Departamento de Patologia, Programa de Pós-graduação em Anatomia Patológica, Faculdade de Medicina, Hospital Universitário Clementina Fraga Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Felipe Leite de Oliveira
- Laboratório de Interações Celulares, Instituto de Ciências Biomédicas, Programa de Pós-graduação em Ciências Morfológicas, Centro de Ciências da Saúde, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Valeria de Mello Coelho
- Laboratório de lmunofisiologia, Instituto de Ciências Biomédicas, Programa de Pós-graduação em Ciências Morfológicas, Centro de Ciências da Saúde, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Emanuela Bezerra Dos Santos Ribeiro
- Laboratório de Neurodegeneração e Reparo, Departamento de Patologia, Programa de Pós-graduação em Anatomia Patológica, Faculdade de Medicina, Hospital Universitário Clementina Fraga Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Juliana Silva de Almeida
- Laboratório de Neurodegeneração e Reparo, Departamento de Patologia, Programa de Pós-graduação em Anatomia Patológica, Faculdade de Medicina, Hospital Universitário Clementina Fraga Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Natalia Moraes Bechelli Silva
- Laboratório de Neurodegeneração e Reparo, Departamento de Patologia, Programa de Pós-graduação em Anatomia Patológica, Faculdade de Medicina, Hospital Universitário Clementina Fraga Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernanda Martins Almeida
- Laboratório de Neurodegeneração e Reparo, Instituto de Ciências Biomédicas, Programa de Pós-graduação em Anatomia Patológica, Centro de Ciências da Saúde, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Maria Blanco Martinez
- Laboratório de Neurodegeneração e Reparo, Departamento de Patologia, Programa de Pós-graduação em Anatomia Patológica, Faculdade de Medicina, Hospital Universitário Clementina Fraga Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
3
|
Guo J, Xu Z, Gunderson RC, Xu B, Michie SA. LFA-1/ICAM-1 Adhesion Pathway Mediates the Homeostatic Migration of Lymphocytes from Peripheral Tissues into Lymph Nodes through Lymphatic Vessels. Biomolecules 2023; 13:1194. [PMID: 37627259 PMCID: PMC10452152 DOI: 10.3390/biom13081194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/22/2023] [Accepted: 07/29/2023] [Indexed: 08/27/2023] Open
Abstract
Lymphocyte function-associated antigen-1 (LFA-1) and its endothelial ligand intercellular adhesion molecule-1 (ICAM-1) are important for the migration of lymphocytes from blood vessels into lymph nodes. However, it is largely unknown whether these molecules mediate the homeostatic migration of lymphocytes from peripheral tissues into lymph nodes through lymphatic vessels. In this study, we find that, in naive mice, ICAM-1 is expressed on the sinus endothelia of lymph nodes, but not on the lymphatic vessels of peripheral tissues. In in vivo lymphocyte migration assays, memory CD4+ T cells migrated to lymph nodes from peripheral tissues much more efficiently than from blood vessels, as compared to naive CD4+ T cells. Moreover, ICAM-1 deficiency in host mice significantly inhibited the migration of adoptively transferred wild-type donor lymphocytes from peripheral tissues, but not from blood vessels, into lymph nodes. The migration of LFA-1-deficient donor lymphocytes from peripheral tissues into the lymph nodes of wild-type host mice was also significantly reduced as compared to wild-type donor lymphocytes. Furthermore, the number of memory T cells in lymph nodes was significantly reduced in the absence of ICAM-1 or LFA-1. Thus, our study extends the functions of the LFA-1/ICAM-1 adhesion pathway, indicating its novel role in controlling the homeostatic migration of lymphocytes from peripheral tissues into lymph nodes and maintaining memory T cellularity in lymph nodes.
Collapse
Affiliation(s)
- Jia Guo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; (J.G.); (Z.X.); (R.C.G.)
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Center for Hypertension Care, Shanxi Medical University First Hospital, Taiyuan 030012, China
| | - Zeyu Xu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; (J.G.); (Z.X.); (R.C.G.)
- Department of Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45219, USA
| | - Rachel C. Gunderson
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; (J.G.); (Z.X.); (R.C.G.)
| | - Baohui Xu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; (J.G.); (Z.X.); (R.C.G.)
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sara A. Michie
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; (J.G.); (Z.X.); (R.C.G.)
| |
Collapse
|
4
|
Bao N, Fu B, Zhong X, Jia S, Ren Z, Wang H, Wang W, Shi H, Li J, Ge F, Chang Q, Gong Y, Liu W, Qiu F, Xu S, Li T. Role of the CXCR6/CXCL16 axis in autoimmune diseases. Int Immunopharmacol 2023; 121:110530. [PMID: 37348231 DOI: 10.1016/j.intimp.2023.110530] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 06/01/2023] [Accepted: 06/15/2023] [Indexed: 06/24/2023]
Abstract
The C-X-C motif ligand 16, or CXCL16, is a chemokine that belongs to the ELR - CXC subfamily. Its function is to bind to the chemokine receptor CXCR6, which is a G protein-coupled receptor with 7 transmembrane domains. The CXCR6/CXCL16 axis has been linked to the development of numerous autoimmune diseases and is connected to clinical parameters that reflect disease severity, activity, and prognosis in conditions such as multiple sclerosis, autoimmune hepatitis, rheumatoid arthritis, Crohn's disease, and psoriasis. CXCL16 is expressed in various immune cells, such as dendritic cells, monocytes, macrophages, and B cells. During autoimmune diseases, CXCL16 can facilitate the adhesion of immune cells like monocytes, T cells, NKT cells, and others to endothelial cells and dendritic cells. Additionally, sCXCL16 can regulate the migration of CXCR6-expressing leukocytes, which includes CD8+ T cells, CD4+ T cells, NK cells, constant natural killer T cells, plasma cells, and monocytes. Further investigation is required to comprehend the intricate interactions between chemokines and the pathogenesis of autoimmune diseases. It remains to be seen whether the CXCR6/CXCL16 axis represents a new target for the treatment of these conditions.
Collapse
Affiliation(s)
- Nandi Bao
- Senior Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Bo Fu
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Beijing, China; State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, China
| | - Xiaoling Zhong
- Department of neurology, School of Medicine, South China University of Technology, Guangzhou, China; Department of neurology, The Sixth Medical Center of PLA General Hospital of Beijing, Beijing, China
| | - Shuangshuang Jia
- Department of neurology, The Sixth Medical Center of PLA General Hospital of Beijing, Beijing, China; Navy Clinical College, the Fifth School of Clinical Medicine, Anhui Medical University, Hefei, China
| | - Zhuangzhuang Ren
- Navy Clinical College, the Fifth School of Clinical Medicine, Anhui Medical University, Hefei, China
| | - Haoran Wang
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Beijing, China; State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, China
| | - Weihua Wang
- Department of Gastroenterology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Hui Shi
- Department of Gastroenterology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Jun Li
- Department of Gastroenterology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Fulin Ge
- Department of Gastroenterology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Qing Chang
- Department of Gastroenterology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Yuan Gong
- Department of Gastroenterology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Wenhui Liu
- Department of Gastroenterology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Feng Qiu
- Senior Department of Neurology, The First Medical Center of PLA General Hospital, Beijing, China.
| | - Shiping Xu
- Department of Gastroenterology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China.
| | - Tingting Li
- State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, China; Department of Gastroenterology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
5
|
Washburn RL, Martinez-Marin D, Sniegowski T, Korać K, Rodriguez AR, Miranda JM, Chilton BS, Bright RK, Pruitt K, Bhutia YD, Dufour JM. Sertoli Cells Express Accommodation, Survival, and Immunoregulatory Factors When Exposed to Normal Human Serum. Biomedicines 2023; 11:1650. [PMID: 37371745 DOI: 10.3390/biomedicines11061650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 05/30/2023] [Accepted: 06/03/2023] [Indexed: 06/29/2023] Open
Abstract
Transplantation is a clinical procedure that treats a variety of diseases yet is unattainable for many patients due to a nationwide organ shortage and the harsh side effects of chronic immune suppression. Xenografted pig organs are an attractive alternative to traditional allografts and would provide an endless supply of transplantable tissue, but transplants risk rejection by the recipient's immune system. An essential component of the rejection immune response is the complement system. Sertoli cells, an immunoregulatory testicular cell, survive complement as xenografts long term without any immune suppressants. We hypothesized that exposure to the xenogeneic complement influences Sertoli cell gene expression of other accommodation factors that contribute to their survival; thus, the purpose of this study was to describe these potential changes in gene expression. RNA sequencing of baseline neonatal pig Sertoli cells (NPSC) as compared to NPSC after exposure to normal human serum (NHS, containing complement) revealed 62 significantly differentially expressed genes (DEG) that affect over 30 pathways involved in immune regulation, cell survival, and transplant accommodation. Twelve genes of interest were selected for further study, and Sertoli cell protein expression of CCL2 and the accommodation factor A20 were confirmed for the first time. Functional pathway analyses were conducted in NPSC and three biological clusters were revealed as being considerably affected by NHS exposure: innate immune signaling, cytokine signaling, and T cell regulation. Better understanding of the interaction of Sertoli cells with complement in a xenograft environment may reveal the mechanisms behind immune-privileged systems to increase graft viability.
Collapse
Affiliation(s)
- Rachel L Washburn
- Department of Cell Biology and Biochemistry, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79424, USA
- Department of Immunology and Molecular Microbiology, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79424, USA
| | - Dalia Martinez-Marin
- Department of Cell Biology and Biochemistry, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79424, USA
- Department of Immunology and Molecular Microbiology, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79424, USA
| | - Tyler Sniegowski
- Department of Cell Biology and Biochemistry, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79424, USA
| | - Ksenija Korać
- Department of Cell Biology and Biochemistry, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79424, USA
| | - Alexis R Rodriguez
- Department of Cell Biology and Biochemistry, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79424, USA
| | - Jonathan M Miranda
- Department of Cell Biology and Biochemistry, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79424, USA
| | - Beverly S Chilton
- Department of Cell Biology and Biochemistry, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79424, USA
| | - Robert K Bright
- Department of Immunology and Molecular Microbiology, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79424, USA
| | - Kevin Pruitt
- Department of Immunology and Molecular Microbiology, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79424, USA
| | - Yangzom D Bhutia
- Department of Cell Biology and Biochemistry, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79424, USA
| | - Jannette M Dufour
- Department of Cell Biology and Biochemistry, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79424, USA
| |
Collapse
|
6
|
Yoshihara T, Okabe Y. Aldh1a2 + fibroblastic reticular cells regulate lymphocyte recruitment in omental milky spots. J Exp Med 2023; 220:213908. [PMID: 36880532 PMCID: PMC9997506 DOI: 10.1084/jem.20221813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 12/29/2022] [Accepted: 02/03/2023] [Indexed: 03/06/2023] Open
Abstract
Lymphoid clusters in visceral adipose tissue omentum, known as milky spots, play a central role in the immunological defense in the abdomen. Milky spots exhibit hybrid nature between secondary lymph organs and ectopic lymphoid tissues, yet their development and maturation mechanisms are poorly understood. Here, we identified a subset of fibroblastic reticular cells (FRCs) that are uniquely present in omental milky spots. These FRCs were characterized by the expression of retinoic acid-converting enzyme, Aldh1a2, and endothelial cell marker, Tie2, in addition to canonical FRC-associated genes. Diphtheria toxin-mediated ablation of Aldh1a2+ FRCs resulted in the alteration in milky spot structure with a significant reduction in size and cellularity. Mechanistically, Aldh1a2+ FRCs regulated the display of chemokine CXCL12 on high endothelial venules (HEVs), which recruit blood-borne lymphocytes from circulation. We further found that Aldh1a2+ FRCs are required for the maintenance of peritoneal lymphocyte composition. These results illustrate the homeostatic roles of FRCs in the formation of non-classical lymphoid tissues.
Collapse
Affiliation(s)
- Tomomi Yoshihara
- Laboratory of Immune Homeostasis, WPI Immunology Frontier Research Center, Osaka University , Osaka, Japan
| | - Yasutaka Okabe
- Laboratory of Immune Homeostasis, WPI Immunology Frontier Research Center, Osaka University , Osaka, Japan.,Center for Infectious Disease Education and Research, Osaka University , Osaka, Japan.,Japan Science and Technology Agency , PRESTO, Kawaguchi, Japan
| |
Collapse
|
7
|
Sîrbe C, Badii M, Crişan TO, Bența G, Grama A, Joosten LAB, Rednic S, Pop TL. Detection of Novel Biomarkers in Pediatric Autoimmune Hepatitis by Proteomic Profiling. Int J Mol Sci 2023; 24:ijms24087479. [PMID: 37108648 PMCID: PMC10141667 DOI: 10.3390/ijms24087479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/14/2023] [Accepted: 04/16/2023] [Indexed: 04/29/2023] Open
Abstract
Autoimmune hepatitis (AIH) is characterized by immune-mediated hepatocyte injury resulting in the destruction of liver cells, causing inflammation, liver failure, and fibrosis. Pediatric (AIH) is an autoimmune inflammatory disease that usually requires immunosuppression for an extended period. Frequent relapses after treatment discontinuation demonstrate that current therapies do not control intrahepatic immune processes. This study describes targeted proteomic profiling data in patients with AIH and controls. A total of 92 inflammatory and 92 cardiometabolic plasma markers were assessed for (i) pediatric AIH versus controls, (ii) AIH type 1 versus type 2, (iii) AIH and AIH-autoimmune sclerosing cholangitis overlapping syndrome and (iv) correlations with circulating vitamin D levels in AIH. A total of 16 proteins showed a nominally significant differential abundance in pediatric patients with AIH compared to controls. No clustering of AIH subphenotypes based on all protein data was observed, and no significant correlation of vitamin D levels was observed for the identified proteins. The proteins that showed variable expression include CA1, CA3, GAS6, FCGR2A, 4E-BP1 and CCL19, which may serve as potential biomarkers for patients with AIH. CX3CL1, CXCL10, CCL23, CSF1 and CCL19 showed homology to one another and may be coexpressed in AIH. CXCL10 seems to be the central intermediary link for the listed proteins. These proteins were involved in relevant mechanistic pathways for liver diseases and immune processes in AIH pathogenesis. This is the first report on the proteomic profile of pediatric AIH. The identified markers could potentially lead to new diagnostic and therapeutic tools. Nevertheless, considering the complex pathogenesis of AIH, more extensive studies are warranted to replicate and validate the present study's findings.
Collapse
Affiliation(s)
- Claudia Sîrbe
- 2nd Pediatric Discipline, Department of Mother and Child, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- 2nd Pediatric Clinic, Center of Expertise in Pediatric Liver Rare Disorders, Emergency Clinical Hospital for Children, 400177 Cluj-Napoca, Romania
| | - Medeea Badii
- Department of Medical Genetics, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Internal Medicine, Radboud University Medical Centre, 6525 XZ Nijmegen, The Netherlands
| | - Tania O Crişan
- Department of Medical Genetics, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Internal Medicine, Radboud University Medical Centre, 6525 XZ Nijmegen, The Netherlands
| | - Gabriel Bența
- 2nd Pediatric Discipline, Department of Mother and Child, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- 2nd Pediatric Clinic, Center of Expertise in Pediatric Liver Rare Disorders, Emergency Clinical Hospital for Children, 400177 Cluj-Napoca, Romania
| | - Alina Grama
- 2nd Pediatric Discipline, Department of Mother and Child, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- 2nd Pediatric Clinic, Center of Expertise in Pediatric Liver Rare Disorders, Emergency Clinical Hospital for Children, 400177 Cluj-Napoca, Romania
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud University Medical Centre, 6525 XZ Nijmegen, The Netherlands
| | - Simona Rednic
- Rheumatology Department, Emergency County Hospital Cluj, 400347 Cluj-Napoca, Romania
- Rheumatology Discipline, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Tudor Lucian Pop
- 2nd Pediatric Discipline, Department of Mother and Child, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- 2nd Pediatric Clinic, Center of Expertise in Pediatric Liver Rare Disorders, Emergency Clinical Hospital for Children, 400177 Cluj-Napoca, Romania
| |
Collapse
|
8
|
Uhl B, Prochazka KT, Pansy K, Wenzl K, Strobl J, Baumgartner C, Szmyra MM, Waha JE, Wolf A, Tomazic PV, Steinbauer E, Steinwender M, Friedl S, Weniger M, Küppers R, Pichler M, Greinix HT, Stary G, Ramsay AG, Apollonio B, Feichtinger J, Beham-Schmid C, Neumeister P, Deutsch AJ. Distinct Chemokine Receptor Expression Profiles in De Novo DLBCL, Transformed Follicular Lymphoma, Richter's Trans-Formed DLBCL and Germinal Center B-Cells. Int J Mol Sci 2022; 23:7874. [PMID: 35887224 PMCID: PMC9316992 DOI: 10.3390/ijms23147874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 11/17/2022] Open
Abstract
Chemokine receptors and their ligands have been identified as playing an important role in the development of diffuse large B-cell lymphoma (DLBCL), follicular lymphoma, and Richter syndrome (RS). Our aim was to investigate the different expression profiles in de novo DLBCL, transformed follicular lymphoma (tFL), and RS. Here, we profiled the mRNA expression levels of 18 chemokine receptors (CCR1-CCR9, CXCR1-CXCR7, CX3CR1 and XCR1) using RQ-PCR, as well as immunohistochemistry of seven chemokine receptors (CCR1, CCR4-CCR8 and CXCR2) in RS, de novo DLBCL, and tFL biopsy-derived tissues. Tonsil-derived germinal center B-cells (GC-B) served as non-neoplastic controls. The chemokine receptor expression profiles of de novo DLBCL and tFL substantially differed from those of GC-B, with at least 5-fold higher expression of 15 out of the 18 investigated chemokine receptors (CCR1-CCR9, CXCR1, CXCR2, CXCR6, CXCR7, CX3CR1 and XCR1) in these lymphoma subtypes. Interestingly, the de novo DLBCL and tFL exhibited at least 22-fold higher expression of CCR1, CCR5, CCR8, and CXCR6 compared with RS, whereas no significant difference in chemokine receptor expression profile was detected when comparing de novo DLBCL with tFL. Furthermore, in de novo DLBCL and tFLs, a high expression of CCR7 was associated with a poor overall survival in our study cohort, as well as in an independent patient cohort. Our data indicate that the chemokine receptor expression profile of RS differs substantially from that of de novo DLBCL and tFL. Thus, these multiple dysregulated chemokine receptors could represent novel clinical markers as diagnostic and prognostic tools. Moreover, this study highlights the relevance of chemokine signaling crosstalk in the tumor microenvironment of aggressive lymphomas.
Collapse
Affiliation(s)
- Barbara Uhl
- Division of Hematology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria; (B.U.); (K.T.P.); (K.P.); (K.W.); (M.M.S.); (H.T.G.)
| | - Katharina T. Prochazka
- Division of Hematology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria; (B.U.); (K.T.P.); (K.P.); (K.W.); (M.M.S.); (H.T.G.)
| | - Katrin Pansy
- Division of Hematology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria; (B.U.); (K.T.P.); (K.P.); (K.W.); (M.M.S.); (H.T.G.)
| | - Kerstin Wenzl
- Division of Hematology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria; (B.U.); (K.T.P.); (K.P.); (K.W.); (M.M.S.); (H.T.G.)
- Division of Hematology, Mayo Clinic, Rochester, MN 55902, USA
| | - Johanna Strobl
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria; (J.S.); (G.S.)
| | - Claudia Baumgartner
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, 8036 Graz, Austria; (C.B.); (J.F.)
| | - Marta M. Szmyra
- Division of Hematology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria; (B.U.); (K.T.P.); (K.P.); (K.W.); (M.M.S.); (H.T.G.)
| | - James E. Waha
- General, Visceral and Transplant Surgery, Medical University of Graz, 8036 Graz, Austria;
| | - Axel Wolf
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Graz, 8036 Graz, Austria; (A.W.); (P.V.T.)
| | - Peter V. Tomazic
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Graz, 8036 Graz, Austria; (A.W.); (P.V.T.)
| | - Elisabeth Steinbauer
- Institute of Pathology, Medical University of Graz, 8036 Graz, Austria; (E.S.); (M.S.); (S.F.); (C.B.-S.)
| | - Maria Steinwender
- Institute of Pathology, Medical University of Graz, 8036 Graz, Austria; (E.S.); (M.S.); (S.F.); (C.B.-S.)
| | - Sabine Friedl
- Institute of Pathology, Medical University of Graz, 8036 Graz, Austria; (E.S.); (M.S.); (S.F.); (C.B.-S.)
| | - Marc Weniger
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, 45122 Essen, Germany; (M.W.); (R.K.)
| | - Ralf Küppers
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, 45122 Essen, Germany; (M.W.); (R.K.)
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Martin Pichler
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria;
| | - Hildegard T. Greinix
- Division of Hematology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria; (B.U.); (K.T.P.); (K.P.); (K.W.); (M.M.S.); (H.T.G.)
| | - Georg Stary
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria; (J.S.); (G.S.)
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, 1090 Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Alan G. Ramsay
- Faculty of Life Sciences & Medicine, School of Cancer & Pharmaceutical Sciences, King’s College London, London WC2R 2LS, UK; (A.G.R.); (B.A.)
| | - Benedetta Apollonio
- Faculty of Life Sciences & Medicine, School of Cancer & Pharmaceutical Sciences, King’s College London, London WC2R 2LS, UK; (A.G.R.); (B.A.)
| | - Julia Feichtinger
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, 8036 Graz, Austria; (C.B.); (J.F.)
| | - Christine Beham-Schmid
- Institute of Pathology, Medical University of Graz, 8036 Graz, Austria; (E.S.); (M.S.); (S.F.); (C.B.-S.)
| | - Peter Neumeister
- Division of Hematology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria; (B.U.); (K.T.P.); (K.P.); (K.W.); (M.M.S.); (H.T.G.)
| | - Alexander J. Deutsch
- Division of Hematology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria; (B.U.); (K.T.P.); (K.P.); (K.W.); (M.M.S.); (H.T.G.)
| |
Collapse
|
9
|
Banerjee S, Nara R, Chakraborty S, Chowdhury D, Haldar S. Integrin Regulated Autoimmune Disorders: Understanding the Role of Mechanical Force in Autoimmunity. Front Cell Dev Biol 2022; 10:852878. [PMID: 35372360 PMCID: PMC8971850 DOI: 10.3389/fcell.2022.852878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 02/08/2022] [Indexed: 11/13/2022] Open
Abstract
The pathophysiology of autoimmune disorders is multifactorial, where immune cell migration, adhesion, and lymphocyte activation play crucial roles in its progression. These immune processes are majorly regulated by adhesion molecules at cell–extracellular matrix (ECM) and cell–cell junctions. Integrin, a transmembrane focal adhesion protein, plays an indispensable role in these immune cell mechanisms. Notably, integrin is regulated by mechanical force and exhibit bidirectional force transmission from both the ECM and cytosol, regulating the immune processes. Recently, integrin mechanosensitivity has been reported in different immune cell processes; however, the underlying mechanics of these integrin-mediated mechanical processes in autoimmunity still remains elusive. In this review, we have discussed how integrin-mediated mechanotransduction could be a linchpin factor in the causation and progression of autoimmune disorders. We have provided an insight into how tissue stiffness exhibits a positive correlation with the autoimmune diseases’ prevalence. This provides a plausible connection between mechanical load and autoimmunity. Overall, gaining insight into the role of mechanical force in diverse immune cell processes and their dysregulation during autoimmune disorders will open a new horizon to understand this physiological anomaly.
Collapse
|
10
|
Lee GY, Kim WK, No JS, Yi Y, Park HC, Jung J, Cho S, Lee J, Lee SH, Park K, Kim J, Song JW. Clinical and Immunological Predictors of Hemorrhagic Fever with Renal Syndrome Outcome during the Early Phase. Viruses 2022; 14:595. [PMID: 35337004 PMCID: PMC8954228 DOI: 10.3390/v14030595] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/04/2022] [Accepted: 03/08/2022] [Indexed: 11/16/2022] Open
Abstract
The ability to accurately predict the early progression of hemorrhagic fever with renal syndrome (HFRS) is crucial for reducing morbidity and mortality rates in severely affected patients. However, the utility of biomarkers for predicting clinical outcomes remains elusive in HFRS. The aims of the current study were to analyze the serum levels of immune function-related proteins and identify novel biomarkers that may help ascertain clinical outcomes of HFRS. Enzyme-linked immunosorbent assay, Luminex, and bioanalyzer assays were used to quantitatively detect 15 biomarkers in 49 serum samples of 26 patients with HFRS. High hemoglobin (HGB) and low urine output (UO) levels were identified as potential biomarkers associated with the acute HFRS. The serum soluble urokinase plasminogen activator receptor (suPAR) and C-X-C motif chemokine ligand 10 (CXCL10) values increased in the early phase of diseases. Elevated suPAR, interleukin-10 (IL-10), CXCL10, and decreased transforming growth factor-beta 3 (TGF-β3) were representative predictors of the disease severity. Upregulation of the HGB showed a significant correlation with high levels of suPAR and CXCL10. Reduced UO positively correlated with increased suPAR, CXCL10, and TGF-β2, and decreased vascular endothelial growth factor and TGF-β3. The changing HGB and UO criteria, high suPAR, IL-10, CXCL10, and low TGF-β3 of HFRS raise significant awareness for physicians regarding prospective biomarkers for monitoring early warning signs of HFRS. This study provides critical insights into the clinical and immunological biomarkers for disease severity and progression in patients with HFRS to identify early predictions of fatal outcomes.
Collapse
Affiliation(s)
- Geum-Young Lee
- Department of Microbiology, College of Medicine, Korea University, Seoul 02841, Korea; (G.-Y.L.); (S.C.); (J.L.); (K.P.); (J.K.)
| | - Won-Keun Kim
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon 24252, Korea;
- Institute of Medical Science, College of Medicine, Hallym University, Chuncheon 24252, Korea
| | - Jin Sun No
- Division of High-Risk Pathogens, Bureau of Infectious Diseases Diagnosis Control, Korea Disease Control and Prevention Agency, Cheongju 28159, Korea;
| | - Yongjin Yi
- Division of Nephrology, Department of Internal Medicine, Dankook University Hospital, Cheonan 31116, Korea;
| | - Hayne Cho Park
- Department of Internal Medicine, Kangnam Sacred Heart Hospital, Seoul 02841, Korea;
| | - Jaehun Jung
- Department of Preventive Medicine, Gachon University College of Medicine, Incheon 21556, Korea;
| | - Seungchan Cho
- Department of Microbiology, College of Medicine, Korea University, Seoul 02841, Korea; (G.-Y.L.); (S.C.); (J.L.); (K.P.); (J.K.)
| | - Jingyeong Lee
- Department of Microbiology, College of Medicine, Korea University, Seoul 02841, Korea; (G.-Y.L.); (S.C.); (J.L.); (K.P.); (J.K.)
| | - Seung-Ho Lee
- Chem-Bio Technology Center, Agency for Defense Development, Daejeon 34075, Korea;
| | - Kyungmin Park
- Department of Microbiology, College of Medicine, Korea University, Seoul 02841, Korea; (G.-Y.L.); (S.C.); (J.L.); (K.P.); (J.K.)
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Korea
| | - Jongwoo Kim
- Department of Microbiology, College of Medicine, Korea University, Seoul 02841, Korea; (G.-Y.L.); (S.C.); (J.L.); (K.P.); (J.K.)
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Korea
| | - Jin-Won Song
- Department of Microbiology, College of Medicine, Korea University, Seoul 02841, Korea; (G.-Y.L.); (S.C.); (J.L.); (K.P.); (J.K.)
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Korea
| |
Collapse
|
11
|
Kottom TJ, Carmona EM, Limper AH. Gene Expression in Lung Epithelial Cells Following Interaction with Pneumocystis carinii and its Specific Life Forms Yields Insights into Host Gene Responses to Infection. Microbiol Immunol 2022; 66:238-251. [PMID: 35229348 PMCID: PMC9090966 DOI: 10.1111/1348-0421.12972] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 02/01/2022] [Accepted: 02/22/2022] [Indexed: 11/29/2022]
Abstract
Pneumocystis spp. interacts with epithelial cells in the alveolar spaces of the lung. It is thought that the binding of Pneumocystis to host cell epithelium is needed for life cycle completion and proliferation. The effect of this interaction on lung epithelial cells have previously shown that the trophic form of this organism greatly inhibits p34 cdc2 activity, a serine/threonine kinase required for transition from G2 to M phase in the cell cycle. To gain further insight into the host response during Pneumocystis pneumonia (PCP), we used microarray technology to profile epithelial cell (A549) gene expression patterns following Pneumocystis carinii interaction. Furthermore, we isolated separate populations of cyst and trophic forms of P. carinii, which were then applied to the lung epithelial cells. Differential expression of genes involved in various cellular functions dependent on the specific P. carinii life form in contact with the A549 cell were identified. The reliability of our data was further confirmed by Northern blot analysis on a number of selected up or down regulated transcripts. The transcriptional response to P. carinii was dominated by cytokines, apoptotic, and anti-apoptotic related genes. These results reveal several previously unknown effects of P. carinii on the lung epithelial cell and provide insight into the complex interactions of host and pathogen. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Theodore J Kottom
- Thoracic Diseases Research Unit, Departments of Medicine and Biochemistry, Mayo Clinic College of Medicine, Rochester, Minnesota, 55905, USA
| | - Eva M Carmona
- Thoracic Diseases Research Unit, Departments of Medicine and Biochemistry, Mayo Clinic College of Medicine, Rochester, Minnesota, 55905, USA
| | - Andrew H Limper
- Thoracic Diseases Research Unit, Departments of Medicine and Biochemistry, Mayo Clinic College of Medicine, Rochester, Minnesota, 55905, USA
| |
Collapse
|
12
|
Chandra A, Butler MT, Bear JE, Haugh JM. Modeling cell protrusion predicts how myosin II and actin turnover affect adhesion-based signaling. Biophys J 2022; 121:102-118. [PMID: 34861242 PMCID: PMC8758409 DOI: 10.1016/j.bpj.2021.11.2889] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/03/2021] [Accepted: 11/29/2021] [Indexed: 01/07/2023] Open
Abstract
Orchestration of cell migration is essential for development, tissue regeneration, and the immune response. This dynamic process integrates adhesion, signaling, and cytoskeletal subprocesses across spatial and temporal scales. In mesenchymal cells, adhesion complexes bound to extracellular matrix mediate both biochemical signal transduction and physical interaction with the F-actin cytoskeleton. Here, we present a mathematical model that offers insight into both aspects, considering spatiotemporal dynamics of nascent adhesions, active signaling molecules, mechanical clutching, actin treadmilling, and nonmuscle myosin II contractility. At the core of the model is a positive feedback loop, whereby adhesion-based signaling promotes generation of barbed ends at, and protrusion of, the cell's leading edge, which in turn promotes formation and stabilization of nascent adhesions. The model predicts a switch-like transition and optimality of membrane protrusion, determined by the balance of actin polymerization and retrograde flow, with respect to extracellular matrix density. The model, together with new experimental measurements, explains how protrusion can be modulated by mechanical effects (nonmuscle myosin II contractility and adhesive bond stiffness) and F-actin turnover.
Collapse
Affiliation(s)
- Ankit Chandra
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina
| | - Mitchell T Butler
- Department of Cell Biology and Physiology, UNC Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - James E Bear
- Department of Cell Biology and Physiology, UNC Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Jason M Haugh
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina.
| |
Collapse
|
13
|
Freff J, Bröker L, Leite Dantas R, Schwarte K, Bühlmeier J, Kraft I, Hinney A, Buhlmann U, Arolt V, Dannlowski U, Romer G, Baune BT, Hebebrand J, Föcker M, Alferink J. Expression of CXCR4 on CD4 + T cells predicts body composition parameters in female adolescents with anorexia nervosa. Front Psychiatry 2022; 13:960905. [PMID: 36226111 PMCID: PMC9549152 DOI: 10.3389/fpsyt.2022.960905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/06/2022] [Indexed: 11/30/2022] Open
Abstract
Anorexia nervosa (AN) is a severe eating disorder characterized by excessive weight loss and lack of recognition of the seriousness of the current low body weight. Individuals with AN frequently exhibit an enhanced inflammatory state and altered blood levels of cytokines and chemokines. However, the expression of chemokine receptors in AN and the association with body composition parameters and treatment effects are still unknown. In this study, we examined the expression of CCR4, CCR6, CXCR3, and CXCR4 on peripheral blood T cells in female adolescents with AN before (T0, n = 24) and after 6 weeks of multimodal therapy (T1, n = 20). We also investigated their value to predict body mass index (BMI) and fat mass index (FMI) at baseline. Using multi-parameter flow cytometry, we found increased expression of CCR4, CXCR3, and CXCR4, but not CCR6, on CD4+ T cells in AN at T0 when compared to healthy controls (HC, n = 20). At T1, CXCR3 and CXCR4 expression decreased in AN. We found a close link between CCR4, CCR6 and CXCR4 expression and the adolescent mental health status in the study cohort as determined by the Strengths and Difficulties Questionnaire (SDQ). Specifically, CXCR4 expression correlated positively with emotional symptoms and peer relationship problems, as well as with the total sum score of the SDQ. In addition, CXCR4 expression on CD4+ T cells was a significant predictor of BMI and FMI in female adolescents. Our findings that CXCR4 expression on T cells is altered in adolescents with AN and predicts body composition parameters in adolescents suggest an impact of this chemokine receptor in the pathogenesis of AN.
Collapse
Affiliation(s)
- Jana Freff
- Department of Psychiatry, University of Münster, Münster, Germany.,Cells in Motion Interfaculty Cluster, University of Münster, Münster, Germany
| | - Lisa Bröker
- Department of Psychiatry, University of Münster, Münster, Germany.,Cells in Motion Interfaculty Cluster, University of Münster, Münster, Germany
| | - Rafael Leite Dantas
- Department of Psychiatry, University of Münster, Münster, Germany.,Cells in Motion Interfaculty Cluster, University of Münster, Münster, Germany
| | - Kathrin Schwarte
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Judith Bühlmeier
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,Department of Clinical Psychology and Psychotherapy, University of Münster, Münster, Germany.,Faculty of Natural Sciences, Institute of Nutrition, Consumption and Health, University Paderborn, Paderborn, Germany
| | - Isabelle Kraft
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Anke Hinney
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ulrike Buhlmann
- Department of Clinical Psychology and Psychotherapy, University of Münster, Münster, Germany
| | - Volker Arolt
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Udo Dannlowski
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Georg Romer
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital Münster, Münster, Germany
| | - Bernhard T Baune
- Department of Psychiatry, University of Münster, Münster, Germany.,Department of Psychiatry, University of Melbourne, Melbourne, VIC, Australia.,The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - Johannes Hebebrand
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Manuel Föcker
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital Münster, Münster, Germany
| | - Judith Alferink
- Department of Psychiatry, University of Münster, Münster, Germany.,Cells in Motion Interfaculty Cluster, University of Münster, Münster, Germany
| |
Collapse
|
14
|
Gate D, Tapp E, Leventhal O, Shahid M, Nonninger TJ, Yang AC, Strempfl K, Unger MS, Fehlmann T, Oh H, Channappa D, Henderson VW, Keller A, Aigner L, Galasko DR, Davis MM, Poston KL, Wyss-Coray T. CD4 + T cells contribute to neurodegeneration in Lewy body dementia. Science 2021; 374:868-874. [PMID: 34648304 PMCID: PMC9122025 DOI: 10.1126/science.abf7266] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Recent studies indicate that the adaptive immune system plays a role in Lewy body dementia (LBD). However, the mechanism regulating T cell brain homing in LBD is unknown. Here, we observed T cells adjacent to Lewy bodies and dopaminergic neurons in post-mortem LBD brains. Single-cell RNA sequencing of cerebrospinal fluid (CSF) identified upregulated expression of C-X-C Motif Chemokine Receptor 4 (CXCR4) in CD4+ T cells in LBD. CSF protein levels of the CXCR4 ligand, C-X-C Motif Chemokine Ligand 12 (CXCL12) were associated with neuroaxonal damage in LBD. Furthermore, we observed clonal expansion and upregulated Interleukin 17A expression by CD4+ T cells stimulated with a phosphorylated α-synuclein epitope. Thus, CXCR4-CXCL12 signaling may represent a mechanistic target for inhibiting pathological interleukin-17-producing T cell trafficking in LBD. The immune system is implicated in the neurodegenerative process of Lewy body dementia.
Collapse
Affiliation(s)
- David Gate
- Department of Neurology, Northwestern University, Chicago, IL, USA.,Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Emma Tapp
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Olivia Leventhal
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Marian Shahid
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Tim J Nonninger
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Andrew C Yang
- Department of Bioengineering, Stanford University, Stanford, CA, USA.,Chemistry, Engineering, and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA
| | - Katharina Strempfl
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria.,QPS Austria GmbH, Parkring 12, 8074 Grambach, Austria
| | - Michael S Unger
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Tobias Fehlmann
- Chair for Clinical Bioinformatics, Saarland University, Saarbrucken, Germany
| | - Hamilton Oh
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Divya Channappa
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Victor W Henderson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Andreas Keller
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Chair for Clinical Bioinformatics, Saarland University, Saarbrucken, Germany
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Douglas R Galasko
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Mark M Davis
- Department of Microbiology and Immunology, School of Medicine, Stanford University, Stanford, CA, USA.,Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Kathleen L Poston
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.,Chemistry, Engineering, and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA
| |
Collapse
|
15
|
Yoshie O. CCR4 as a Therapeutic Target for Cancer Immunotherapy. Cancers (Basel) 2021; 13:cancers13215542. [PMID: 34771703 PMCID: PMC8583476 DOI: 10.3390/cancers13215542] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/01/2021] [Accepted: 11/03/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary CCR4 is a chemokine receptor selectively expressed on normal T cell subsets such as type 2 helper T cells, skin-homing T cells and regulatory T cells, and on skin-associated T cell malignancies such as adult T cell leukemia/lymphoma (ATLL), which is etiologically associated with human T lymphocyte virus type 1 (HTLV-1), and cutaneous T cell lymphomas (CTCLs). Mogamulizumab is a fully humanized and glyco-engineered monoclonal anti-CCR4 antibody used for the treatment of refractory/relapsed ATLL and CTCLs, often resulting in complete remission. The clinical applications of Mogamulizumab are now being extended to solid tumors, exploring the therapeutic effect of regulatory T cell depletion. This review overviews the expression of CCR4 in various T cell subsets, HTLV-1-infected T cells, ATLL and CTCLs, and the clinical applications of Mogamulizumab. Abstract CCR4 is a chemokine receptor mainly expressed by T cells. It is the receptor for two CC chemokine ligands, CCL17 and CCL22. Originally, the expression of CCR4 was described as highly selective for helper T type 2 (Th2) cells. Later, its expression was extended to other T cell subsets such as regulatory T (Treg) cells and Th17 cells. CCR4 has long been regarded as a potential therapeutic target for allergic diseases such as atopic dermatitis and bronchial asthma. Furthermore, the findings showing that CCR4 is strongly expressed by T cell malignancies such as adult T cell leukemia/lymphoma (ATLL) and cutaneous T cell lymphomas (CTCLs) have led to the development and clinical application of the fully humanized and glyco-engineered monoclonal anti-CCR4 Mogamulizumab in refractory/relapsed ATLL and CTCLs with remarkable successes. However, Mogamulizumab often induces severe adverse events in the skin possibly because of its efficient depletion of Treg cells. In particular, treatment with Mogamulizumab prior to allogenic hematopoietic stem cell transplantation (allo-HSCT), the only curative option of these T cell malignancies, often leads to severe glucocorticoid-refractory graft-versus-host diseases. The efficient depletion of Treg cells by Mogamulizumab has also led to its clinical trials in advanced solid tumors singly or in combination with immune checkpoint inhibitors. The main focus of this review is CCR4; its expression on normal and malignant T cells and its significance as a therapeutic target in cancer immunotherapy.
Collapse
Affiliation(s)
- Osamu Yoshie
- Health and Kampo Institute, Sendai 981-3205, Japan;
- Kindai University, Osaka 577-8502, Japan
- Aoinosono-Sendai Izumi Long-Term Health Care Facility, Sendai 981-3126, Japan
| |
Collapse
|
16
|
Cho E, Theall B, Stampley J, Granger J, Johannsen NM, Irving BA, Spielmann G. Cytomegalovirus Infection Impairs the Mobilization of Tissue-Resident Innate Lymphoid Cells into the Peripheral Blood Compartment in Response to Acute Exercise. Viruses 2021; 13:v13081535. [PMID: 34452400 PMCID: PMC8402764 DOI: 10.3390/v13081535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/24/2021] [Accepted: 07/29/2021] [Indexed: 11/25/2022] Open
Abstract
Circulating immune cell numbers and phenotypes are impacted by high-intensity acute bouts of exercise and infection history with the latent herpesviruses cytomegalovirus (CMV). In particular, CMV infection history impairs the exercise-induced mobilization of cytotoxic innate lymphoid cells 1 (ILC1) cells, also known as NK cells, in the blood. However, it remains unknown whether exercise and CMV infection modulate the mobilization of traditionally tissue-resident non-cytotoxic ILCs into the peripheral blood compartment. To address this question, 22 healthy individuals with or without CMV (20–35 years—45% CMVpos) completed 30 min of cycling at 70% VO2 max, and detailed phenotypic analysis of circulating ILCs was performed at rest and immediately post-exercise. We show for the first time that a bout of high-intensity exercise is associated with an influx of ILCs that are traditionally regarded as tissue-resident. In addition, this is the first study to highlight that latent CMV infection blunts the exercise-response of total ILCs and progenitor ILCs (ILCPs). These promising data suggest that acute exercise facilitates the circulation of certain ILC subsets, further advocating for the improvements in health seen with exercise by enhancing cellular mobilization and immunosurveillance, while also highlighting the indirect deleterious effects of CMV infection in healthy adults.
Collapse
Affiliation(s)
- Eunhan Cho
- School of Kinesiology, Louisiana State University, Baton Rouge, LA 70803, USA; (E.C.); (B.T.); (J.S.); (J.G.); (N.M.J.); (B.A.I.)
| | - Bailey Theall
- School of Kinesiology, Louisiana State University, Baton Rouge, LA 70803, USA; (E.C.); (B.T.); (J.S.); (J.G.); (N.M.J.); (B.A.I.)
| | - James Stampley
- School of Kinesiology, Louisiana State University, Baton Rouge, LA 70803, USA; (E.C.); (B.T.); (J.S.); (J.G.); (N.M.J.); (B.A.I.)
| | - Joshua Granger
- School of Kinesiology, Louisiana State University, Baton Rouge, LA 70803, USA; (E.C.); (B.T.); (J.S.); (J.G.); (N.M.J.); (B.A.I.)
| | - Neil M. Johannsen
- School of Kinesiology, Louisiana State University, Baton Rouge, LA 70803, USA; (E.C.); (B.T.); (J.S.); (J.G.); (N.M.J.); (B.A.I.)
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Brian A. Irving
- School of Kinesiology, Louisiana State University, Baton Rouge, LA 70803, USA; (E.C.); (B.T.); (J.S.); (J.G.); (N.M.J.); (B.A.I.)
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Guillaume Spielmann
- School of Kinesiology, Louisiana State University, Baton Rouge, LA 70803, USA; (E.C.); (B.T.); (J.S.); (J.G.); (N.M.J.); (B.A.I.)
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
- Correspondence:
| |
Collapse
|
17
|
Frutiger A, Tanno A, Hwu S, Tiefenauer RF, Vörös J, Nakatsuka N. Nonspecific Binding-Fundamental Concepts and Consequences for Biosensing Applications. Chem Rev 2021; 121:8095-8160. [PMID: 34105942 DOI: 10.1021/acs.chemrev.1c00044] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Nature achieves differentiation of specific and nonspecific binding in molecular interactions through precise control of biomolecules in space and time. Artificial systems such as biosensors that rely on distinguishing specific molecular binding events in a sea of nonspecific interactions have struggled to overcome this issue. Despite the numerous technological advancements in biosensor technologies, nonspecific binding has remained a critical bottleneck due to the lack of a fundamental understanding of the phenomenon. To date, the identity, cause, and influence of nonspecific binding remain topics of debate within the scientific community. In this review, we discuss the evolution of the concept of nonspecific binding over the past five decades based upon the thermodynamic, intermolecular, and structural perspectives to provide classification frameworks for biomolecular interactions. Further, we introduce various theoretical models that predict the expected behavior of biosensors in physiologically relevant environments to calculate the theoretical detection limit and to optimize sensor performance. We conclude by discussing existing practical approaches to tackle the nonspecific binding challenge in vitro for biosensing platforms and how we can both address and harness nonspecific interactions for in vivo systems.
Collapse
Affiliation(s)
- Andreas Frutiger
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, Zürich CH-8092, Switzerland
| | - Alexander Tanno
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, Zürich CH-8092, Switzerland
| | - Stephanie Hwu
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, Zürich CH-8092, Switzerland
| | - Raphael F Tiefenauer
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, Zürich CH-8092, Switzerland
| | - János Vörös
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, Zürich CH-8092, Switzerland
| | - Nako Nakatsuka
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, Zürich CH-8092, Switzerland
| |
Collapse
|
18
|
Tregs self-organize into a computing ecosystem and implement a sophisticated optimization algorithm for mediating immune response. Proc Natl Acad Sci U S A 2021; 118:2011709118. [PMID: 33372155 DOI: 10.1073/pnas.2011709118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Regulatory T cells (Tregs) play a crucial role in mediating immune response. Yet an algorithmic understanding of the role of Tregs in adaptive immunity remains lacking. Here, we present a biophysically realistic model of Treg-mediated self-tolerance in which Tregs bind to self-antigens and locally inhibit the proliferation of nearby activated T cells. By exploiting a duality between ecological dynamics and constrained optimization, we show that Tregs tile the potential antigen space while simultaneously minimizing the overlap between Treg activation profiles. We find that for sufficiently high Treg diversity, Treg-mediated self-tolerance is robust to fluctuations in self-antigen concentrations but lowering the Treg diversity results in a sharp transition-related to the Gardner transition in perceptrons-to a regime where changes in self-antigen concentrations can result in an autoimmune response. We propose an experimental test of this transition in immune-deficient mice and discuss potential implications for autoimmune diseases.
Collapse
|
19
|
Lu G, Qiu Y, Su X. Targeting CXCL12-CXCR4 Signaling Enhances Immune Checkpoint Blockade Therapy Against Triple Negative Breast Cancer. Eur J Pharm Sci 2021; 157:105606. [PMID: 33131745 DOI: 10.1016/j.ejps.2020.105606] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 06/21/2020] [Accepted: 10/16/2020] [Indexed: 12/18/2022]
Abstract
Insufficient T cell infiltration in triple-negative breast cancer (TNBC) has limited its response rate to immune checkpoint blockade (ICB) therapies and motivated the development of immunostimulatory approaches to enhance the ICB therapy. CXCR4 is a chemokine receptor highly upregulated both on cell surface and cytoplasm in tumor tissues. Activating CXCR4 has been associated with increased immunosuppression in the tumor microenvironment. Here, we developed a CXCR4-targeted liposomal formulation (Liposomal-AMD3100) to enhance therapeutic efficacy of AMD3100, a CXCR4 antagonist. Particularly, AMD3100 is not only encapsulated into the liposome but coated on the surface of the formulation to serve as a targeting moiety and a dual blocker capable of inhibiting CXCR4 activation extracellularly and intracellularly. The Liposomal-AMD3100 remodeled both immune and stromal microenvironment more efficiently compared with free AMD3100, indicating better pharmacodynamic profile of AMD3100 achieved by liposomal formulation. The combination of anti-PD-L1 with Liposomal-AMD3100 formulation exhibited an increased antitumor effect and prolonged survival time compared with monotherapies in a murine TNBC model (4T1). This work proves that immune activation via liposomal delivery of CXCR4 inhibitors has a great potential to expand ICB therapies to originally ICB-insensitive cancer types.
Collapse
Affiliation(s)
- Guowen Lu
- Department of Thyroid and breast mininally invasive surgery, Ningbo Yinzhou People's Hospital, No.251 Baizhang East Road, 315000 Ningbo, Zhejiang, P.R. China.
| | - Yier Qiu
- Department of Thyroid and breast mininally invasive surgery, Ningbo Yinzhou People's Hospital, No.251 Baizhang East Road, 315000 Ningbo, Zhejiang, P.R. China
| | - Xiaobao Su
- Department of Thyroid and breast mininally invasive surgery, Ningbo Yinzhou People's Hospital, No.251 Baizhang East Road, 315000 Ningbo, Zhejiang, P.R. China
| |
Collapse
|
20
|
Shen Y, Zhang Y, Chen L, Du J, Bao H, Xing Y, Cai M, Si Y. Chemokine CXCL13 acts via CXCR5-ERK signaling in hippocampus to induce perioperative neurocognitive disorders in surgically treated mice. J Neuroinflammation 2020; 17:335. [PMID: 33161894 PMCID: PMC7648984 DOI: 10.1186/s12974-020-02013-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 10/28/2020] [Indexed: 12/19/2022] Open
Abstract
Background Perioperative neurocognitive disorders (PNDs) occur frequently after surgery and worsen patient outcome. How C-X-C motif chemokine (CXCL) 13 and its sole receptor CXCR5 contribute to PNDs remains poorly understood. Methods A PND model was created in adult male C57BL/6J and CXCR5−/− mice by exploratory laparotomy. Mice were pretreated via intracerebroventricular injection with recombinant CXCL13, short hairpin RNA against CXCL13 or a scrambled control RNA, or ERK inhibitor PD98059. Then surgery was performed to induce PNDs, and animals were assessed in the Barnes maze trial followed by a fear-conditioning test. Expression of CXCL13, CXCR5, and ERK in hippocampus was examined using Western blot, quantitative PCR, and immunohistochemistry. Levels of interleukin-1 beta (IL-1β) and tumor necrosis factor alpha (TNF-α) in hippocampus were assessed by Western blot. Results Surgery impaired learning and memory, and it increased expression of CXCL13 and CXCR5 in the hippocampus. CXCL13 knockdown partially reversed the effects of surgery on CXCR5 and cognitive dysfunction. CXCR5 knockout led to similar cognitive outcomes as CXCL13 knockdown, and it repressed surgery-induced activation of ERK and production of IL-1β and TNF-α in hippocampus. Recombinant CXCL13 induced cognitive deficits and increased the expression of phospho-ERK as well as IL-1β and TNF-α in hippocampus of wild-type mice, but not CXCR5−/− mice. PD98059 partially blocked CXCL13-induced cognitive dysfunction as well as production of IL-1β and TNF-α. Conclusions CXCL13-induced activation of CXCR5 may contribute to PNDs by triggering ERK-mediated production of pro-inflammatory cytokines in hippocampus.
Collapse
Affiliation(s)
- Yanan Shen
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, People's Republic of China
| | - Yuan Zhang
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, People's Republic of China
| | - Lihai Chen
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, People's Republic of China
| | - Jiayue Du
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, People's Republic of China
| | - Hongguang Bao
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, People's Republic of China
| | - Yan Xing
- Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, School of Mechanical Engineering, Southeast University, Nanjing, 211118, People's Republic of China
| | - Mengmeng Cai
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, People's Republic of China
| | - Yanna Si
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, People's Republic of China.
| |
Collapse
|
21
|
Eberlein J, Davenport B, Nguyen TT, Victorino F, Jhun K, van der Heide V, Kuleshov M, Ma'ayan A, Kedl R, Homann D. Chemokine Signatures of Pathogen-Specific T Cells I: Effector T Cells. THE JOURNAL OF IMMUNOLOGY 2020; 205:2169-2187. [PMID: 32948687 DOI: 10.4049/jimmunol.2000253] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 08/07/2020] [Indexed: 12/16/2022]
Abstract
The choreography of complex immune responses, including the priming, differentiation, and modulation of specific effector T cell populations generated in the immediate wake of an acute pathogen challenge, is in part controlled by chemokines, a large family of mostly secreted molecules involved in chemotaxis and other patho/physiological processes. T cells are both responsive to various chemokine cues and a relevant source for certain chemokines themselves; yet, the actual range, regulation, and role of effector T cell-derived chemokines remains incompletely understood. In this study, using different in vivo mouse models of viral and bacterial infection as well as protective vaccination, we have defined the entire spectrum of chemokines produced by pathogen-specific CD8+ and CD4+T effector cells and delineated several unique properties pertaining to the temporospatial organization of chemokine expression patterns, synthesis and secretion kinetics, and cooperative regulation. Collectively, our results position the "T cell chemokine response" as a notably prominent, largely invariant, yet distinctive force at the forefront of pathogen-specific effector T cell activities and establish novel practical and conceptual approaches that may serve as a foundation for future investigations into the role of T cell-produced chemokines in infectious and other diseases.
Collapse
Affiliation(s)
- Jens Eberlein
- Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Bennett Davenport
- Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Tom T Nguyen
- Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Francisco Victorino
- Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Kevin Jhun
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Verena van der Heide
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Maxim Kuleshov
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029; and.,Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Avi Ma'ayan
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029; and.,Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Ross Kedl
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Dirk Homann
- Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045; .,Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
22
|
Pontes Ferreira C, de Moro Cariste L, Henrique Noronha I, Fernandes Durso D, Lannes-Vieira J, Ramalho Bortoluci K, Araki Ribeiro D, Golenbock D, Gazzinelli RT, de Vasconcelos JRC. CXCR3 chemokine receptor contributes to specific CD8+ T cell activation by pDC during infection with intracellular pathogens. PLoS Negl Trop Dis 2020; 14:e0008414. [PMID: 32574175 PMCID: PMC7337401 DOI: 10.1371/journal.pntd.0008414] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 07/06/2020] [Accepted: 05/22/2020] [Indexed: 11/21/2022] Open
Abstract
Chemokine receptor type 3 (CXCR3) plays an important role in CD8+ T cells migration during intracellular infections, such as Trypanosoma cruzi. In addition to chemotaxis, CXCR3 receptor has been described as important to the interaction between antigen-presenting cells and effector cells. We hypothesized that CXCR3 is fundamental to T. cruzi-specific CD8+ T cell activation, migration and effector function. Anti-CXCR3 neutralizing antibody administration to acutely T. cruzi-infected mice decreased the number of specific CD8+ T cells in the spleen, and those cells had impaired in activation and cytokine production but unaltered proliferative response. In addition, anti-CXCR3-treated mice showed decreased frequency of CD8+ T cells in the heart and numbers of plasmacytoid dendritic cells in spleen and lymph node. As CD8+ T cells interacted with plasmacytoid dendritic cells during infection by T. cruzi, we suggest that anti-CXCR3 treatment lowers the quantity of plasmacytoid dendritic cells, which may contribute to impair the prime of CD8+ T cells. Understanding which molecules and mechanisms guide CD8+ T cell activation and migration might be a key to vaccine development against Chagas disease as those cells play an important role in T. cruzi infection control. Inflammatory chemokine receptors such as CXCR3 play an important role in T lymphocytes migration into an infected tissue during Th1 response. Recently, the role of CXCR3 as a co-stimulatory molecule was demonstrated, and T lymphocytes from CXCR3 deficient mice had impaired effector function. CXCR3 receptor was highly expressed on specific CD8+ T cells after challenge with T. cruzi, and the hypothesis of that molecule is important for CD8+ T cells activation, migration and functionality was raised. We used the anti-CXCR3 neutralizing antibody approach and demonstrated that C57BL/6 treated mice died very quickly due to T. cruzi infection, and specific CD8+ T cells had decreased effector phenotyping, cytokine production, and cytotoxicity. In addition, anti-CXCR3 treatment decreased the number of dendritic plasmacytoid cells in the lymphoid tissues. The lower quantity of dendritic plasmacytoid cells in those tissues might contribute to the decrease in CD8+ T cells activation. Overall, CXCR3 molecule seems to be an important molecule to be explored during vaccine against Chagas disease strategies.
Collapse
Affiliation(s)
- Camila Pontes Ferreira
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
| | | | - Isaú Henrique Noronha
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
| | - Danielle Fernandes Durso
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Joseli Lannes-Vieira
- Laboratory of Biology of the Interactions, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
| | | | - Daniel Araki Ribeiro
- Department of Biosciences of the Federal University of São Paulo, Santos, Brazil
| | - Douglas Golenbock
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Ricardo Tostes Gazzinelli
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - José Ronnie Carvalho de Vasconcelos
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
- Department of Biosciences of the Federal University of São Paulo, Santos, Brazil
- * E-mail:
| |
Collapse
|
23
|
Haines RR, Scharer CD, Lobby JL, Boss JM. LSD1 Cooperates with Noncanonical NF-κB Signaling to Regulate Marginal Zone B Cell Development. THE JOURNAL OF IMMUNOLOGY 2019; 203:1867-1881. [PMID: 31492745 DOI: 10.4049/jimmunol.1900654] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 08/05/2019] [Indexed: 12/25/2022]
Abstract
Marginal zone B cells (MZB) are a mature B cell subset that rapidly respond to blood-borne pathogens. Although the transcriptional changes that occur throughout MZB development are known, the corresponding epigenetic changes and epigenetic modifying proteins that facilitate these changes are poorly understood. The histone demethylase LSD1 is an epigenetic modifier that promotes plasmablast formation, but its role in B cell development has not been explored. In this study, a role for LSD1 in the development of B cell subsets was examined. B cell-conditional deletion of LSD1 in mice resulted in a decrease in MZB whereas follicular B cells and bone marrow B cell populations were minimally affected. LSD1 repressed genes in MZB that were normally upregulated in the myeloid and follicular B cell lineages. Correspondingly, LSD1 regulated chromatin accessibility at the motifs of transcription factors known to regulate splenic B cell development, including NF-κB motifs. The importance of NF-κB signaling was examined through an ex vivo MZB development assay, which showed that both LSD1-deficient and NF-κB-inhibited transitional B cells failed to undergo full MZB development. Gene expression and chromatin accessibility analyses of in vivo- and ex vivo-generated LSD1-deficient MZB indicated that LSD1 regulated the downstream target genes of noncanonical NF-κB signaling. Additionally LSD1 was found to interact with the noncanonical NF-κB transcription factor p52. Together, these data reveal that the epigenetic modulation of the noncanonical NF-κB signaling pathway by LSD1 is an essential process during the development of MZB.
Collapse
Affiliation(s)
- Robert R Haines
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322
| | - Christopher D Scharer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322
| | - Jenna L Lobby
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322
| | - Jeremy M Boss
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322
| |
Collapse
|
24
|
Pontes Ferreira C, Cariste LM, Ferri Moraschi B, Ferrarini Zanetti B, Won Han S, Araki Ribeiro D, Vieira Machado A, Lannes-Vieira J, Gazzinelli RT, Vasconcelos JRC. CXCR3 chemokine receptor guides Trypanosoma cruzi-specific T-cells triggered by DNA/adenovirus ASP2 vaccine to heart tissue after challenge. PLoS Negl Trop Dis 2019; 13:e0007597. [PMID: 31356587 PMCID: PMC6687206 DOI: 10.1371/journal.pntd.0007597] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 08/08/2019] [Accepted: 07/02/2019] [Indexed: 12/15/2022] Open
Abstract
CD8+ T lymphocytes play an important role in controlling infections by intracellular pathogens. Chemokines and their receptors are crucial for the migration of CD8+ T-lymphocytes, which are the main IFNγ producers and cytotoxic effectors cells. Although the participation of chemokine ligands and receptors has been largely explored in viral infection, much less is known in infection by Trypanosoma cruzi, the causative agent of Chagas disease. After T. cruzi infection, CXCR3 chemokine receptor is highly expressed on the surface of CD8+ T-lymphocytes. Here, we hypothesized that CXCR3 is a key molecule for migration of parasite-specific CD8+ T-cells towards infected tissues, where they may play their effector activities. Using a model of induction of resistance to highly susceptible A/Sn mice using an ASP2-carrying DNA/adenovirus prime-boost strategy, we showed that CXCR3 expression was upregulated on CD8+ T-cells, which selectively migrated towards its ligands CXCL9 and CXCL10. Anti-CXCR3 administration reversed the vaccine-induced resistance to T. cruzi infection in a way associated with hampered cytotoxic activity and increased proapoptotic markers on the H2KK-restricted TEWETGQI-specific CD8+ T-cells. Furthermore, CXCR3 receptor critically guided TEWETGQI-specific effector CD8+ T-cells to the infected heart tissue that express CXCL9 and CXCL10. Overall, our study pointed CXCR3 and its ligands as key molecules to drive T. cruzi-specific effector CD8+ T-cells into the infected heart tissue. The unveiling of the process driving cell migration and colonization of infected tissues by pathogen-specific effector T-cells is a crucial requirement to the development of vaccine strategies.
Collapse
Affiliation(s)
- Camila Pontes Ferreira
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
| | | | - Barbara Ferri Moraschi
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
| | | | - Sang Won Han
- Department of Biophysics, Federal University of São Paulo, São Paulo, Brazil
| | | | | | - Joseli Lannes-Vieira
- Laboratory of Biology of the Interactions, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, Brazil
| | - Ricardo Tostes Gazzinelli
- René Rachou Research Center, Fiocruz, Minas Gerais, Brazil
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, United States ofAmerica
| | - José Ronnie Carvalho Vasconcelos
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
- Department of Biosciences, Federal University of São Paulo, Santos, Brazil
- * E-mail:
| |
Collapse
|
25
|
Yossipof TE, Bazak ZR, Kenigsbuch-Sredni D, Caspi RR, Kalechman Y, Sredni B. Tellurium Compounds Prevent and Reverse Type-1 Diabetes in NOD Mice by Modulating α4β7 Integrin Activity, IL-1β, and T Regulatory Cells. Front Immunol 2019; 10:979. [PMID: 31191514 PMCID: PMC6549385 DOI: 10.3389/fimmu.2019.00979] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 04/16/2019] [Indexed: 12/25/2022] Open
Abstract
The study shows that treatment of NOD mice with either of two tellurium-based small molecules, AS101 [ammonium trichloro(dioxoethylene-o,o')tellurate] or SAS [octa-O-bis-(R,R)-tartarate ditellurane] could preserve β cells function and mass. These beneficial effects were reflected in decreased incidence of diabetes, improved glucose clearance, preservation of body weight, and increased survival. The normal glucose levels were associated with increased insulin levels, preservation of β cell mass and increased islet size. Importantly, this protective activity could be demonstrated when the compounds were administered either at the early pre-diabetic phase with no or initial insulitis, at the pre-diabetic stage with advanced insulitis, or even at the advanced, overtly diabetic stage. We further demonstrate that both tellurium compounds prevent migration of autoimmune lymphocytes to the pancreas, via inhibition of the α4β7 integrin activity. Indeed, the decreased migration resulted in diminished pancreatic islets damage both with respect to their size, β cell function, and caspase-3 activity, the hallmark of apoptosis. Most importantly, AS101 and SAS significantly elevated the number of T regulatory cells in the pancreas, thus potentially controlling the autoimmune process. We show that the compounds inhibit pancreatic caspase-1 activity followed by decreased levels of the inflammatory cytokines IL-1β and IL-17 in the pancreas. These properties enable the compounds to increase the proportion of Tregs in the pancreatic lymph nodes. AS101 and SAS have been previously shown to regulate specific integrins through a unique redox mechanism. Our current results suggest that amelioration of disease in NOD mice by this unique mechanism is due to decreased infiltration of pancreatic islets combined with increased immune regulation, leading to decreased inflammation within the islets. As these tellurium compounds show remarkable lack of toxicity in clinical trials (AS101) and pre-clinical studies (SAS), they may be suitable for the treatment of type-1 diabetes.
Collapse
Affiliation(s)
- Tom Eitan Yossipof
- The Mina & Everard Goodman Faculty of Life Sciences, The Safdiè AIDS and Immunology Research Center, C.A.I.R. Institute, Ramat Gan, Israel
| | - Ziva Roy Bazak
- The Mina & Everard Goodman Faculty of Life Sciences, The Safdiè AIDS and Immunology Research Center, C.A.I.R. Institute, Ramat Gan, Israel
| | | | - Rachel R Caspi
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, United States
| | - Yona Kalechman
- The Mina & Everard Goodman Faculty of Life Sciences, The Safdiè AIDS and Immunology Research Center, C.A.I.R. Institute, Ramat Gan, Israel
| | - Benjamin Sredni
- The Mina & Everard Goodman Faculty of Life Sciences, The Safdiè AIDS and Immunology Research Center, C.A.I.R. Institute, Ramat Gan, Israel
| |
Collapse
|
26
|
Ledderose C, Liu K, Kondo Y, Slubowski CJ, Dertnig T, Denicoló S, Arbab M, Hubner J, Konrad K, Fakhari M, Lederer JA, Robson SC, Visner GA, Junger WG. Purinergic P2X4 receptors and mitochondrial ATP production regulate T cell migration. J Clin Invest 2018; 128:3583-3594. [PMID: 29894310 DOI: 10.1172/jci120972] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 05/31/2018] [Indexed: 12/12/2022] Open
Abstract
T cells must migrate in order to encounter antigen-presenting cells (APCs) and to execute their varied functions in immune defense and inflammation. ATP release and autocrine signaling through purinergic receptors contribute to T cell activation at the immune synapse that T cells form with APCs. Here, we show that T cells also require ATP release and purinergic signaling for their migration to APCs. We found that the chemokine stromal-derived factor-1α (SDF-1α) triggered mitochondrial ATP production, rapid bursts of ATP release, and increased migration of primary human CD4+ T cells. This process depended on pannexin-1 ATP release channels and autocrine stimulation of P2X4 receptors. SDF-1α stimulation caused localized accumulation of mitochondria with P2X4 receptors near the front of cells, resulting in a feed-forward signaling mechanism that promotes cellular Ca2+ influx and sustains mitochondrial ATP synthesis at levels needed for pseudopod protrusion, T cell polarization, and cell migration. Inhibition of P2X4 receptors blocked the activation and migration of T cells in vitro. In a mouse lung transplant model, P2X4 receptor antagonist treatment prevented the recruitment of T cells into allograft tissue and the rejection of lung transplants. Our findings suggest that P2X4 receptors are therapeutic targets for immunomodulation in transplantation and inflammatory diseases.
Collapse
Affiliation(s)
- Carola Ledderose
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Kaifeng Liu
- Department of Medicine/Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yutaka Kondo
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Christian J Slubowski
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Thomas Dertnig
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Sara Denicoló
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Mona Arbab
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Johannes Hubner
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Kirstin Konrad
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Mahtab Fakhari
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - James A Lederer
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Simon C Robson
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Gary A Visner
- Department of Medicine/Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Wolfgang G Junger
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.,Ludwig Boltzmann Institute for Traumatology, Vienna, Austria
| |
Collapse
|
27
|
Sun M, Sun L, Sun D, Zhang C, Li M. Targeted delivery of immuno-RNase may improve cancer therapy. Cancer Cell Int 2018; 18:58. [PMID: 29686536 PMCID: PMC5902972 DOI: 10.1186/s12935-018-0546-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 03/22/2018] [Indexed: 11/10/2022] Open
Abstract
Background Immunotoxins are typical therapeutic drugs that can target cancer cells. They exploit the affinity of specific monoclonal antibodies or ligands to cancer cells to deliver a conjugated protein toxin to target sites, thus, attacking the cancer cells. Methods The immuno-RNase, Onc-V3, showed the stability of Onc-V3 in the blood stream. Flow cytometry showed that apoptosis occurred in the HO-8910PM cells when treated with Onc-V3. Under the confocal microscope, the green fluorescent, FITC-Onc-V3, were located in the cytoplasm, suggesting that Onc-V3 had a function in the cytoplasm of cancer cells. Moreover, after staining by DAPI, the blue fluorescent nuclei showed shrinkage and grainy. Wound healing assay showed that high concentrations of Onc-V3 inhibited cell migration and the transwell invasion assay showed that Onc-V3 could inhibit cell invasion to the basement membrane. Western blot results showed significantly decreased PARP, procaspase-9, and procaspase-3 in Onc-V3-induced apoptosis. Results These results of the experiments in vitro had shown that the Onc-V3 could be delivered to the cancer cells accurately and it had strong cytotoxicity on high metastatic cancer cells. Conclusion The specific toxicity of Onc-V3 on highly metastatic cancer cells can make it a promising anti-cancer drug by using V3 to target delivery of Onconase.
Collapse
Affiliation(s)
- Miaonan Sun
- 1Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, 130021 China
| | - Liankun Sun
- 1Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, 130021 China
| | - Dejun Sun
- 2Department of Biomedicine, Regeneration Medicine Institute, College of Pharmacy, Jilin University, Changchun, 130021 China
| | - Chunmei Zhang
- 2Department of Biomedicine, Regeneration Medicine Institute, College of Pharmacy, Jilin University, Changchun, 130021 China
| | - Mei Li
- 2Department of Biomedicine, Regeneration Medicine Institute, College of Pharmacy, Jilin University, Changchun, 130021 China
| |
Collapse
|
28
|
Rot A, Massberg S, Khandoga AG, von Andrian UH. Chemokines and Hematopoietic Cell Trafficking. Hematology 2018. [DOI: 10.1016/b978-0-323-35762-3.00013-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
29
|
Tischner D, Grimm M, Kaur H, Staudenraus D, Carvalho J, Looso M, Günther S, Wanke F, Moos S, Siller N, Breuer J, Schwab N, Zipp F, Waisman A, Kurschus FC, Offermanns S, Wettschureck N. Single-cell profiling reveals GPCR heterogeneity and functional patterning during neuroinflammation. JCI Insight 2017; 2:95063. [PMID: 28768912 DOI: 10.1172/jci.insight.95063] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 06/20/2017] [Indexed: 12/31/2022] Open
Abstract
GPCR expression was intensively studied in bulk cDNA of leukocyte populations, but limited data are available with respect to expression in individual cells. Here, we show a microfluidic-based single-cell GPCR expression analysis in primary T cells, myeloid cells, and endothelial cells under naive conditions and during experimental autoimmune encephalomyelitis, the mouse model of multiple sclerosis. We found that neuroinflammation induces characteristic changes in GPCR heterogeneity and patterning, and we identify various functionally relevant subgroups with specific GPCR profiles among spinal cord-infiltrating CD4 T cells, macrophages, microglia, or endothelial cells. Using GPCRs CXCR4, S1P1, and LPHN2 as examples, we show how this information can be used to develop new strategies for the functional modulation of Th17 cells and activated endothelial cells. Taken together, single-cell GPCR expression analysis identifies functionally relevant subpopulations with specific GPCR repertoires and provides a basis for the development of new therapeutic strategies in immune disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Stefan Günther
- ECCPS Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | | | | | - Nelly Siller
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn2, ), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | | | | | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn2, ), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | | | | | - Stefan Offermanns
- Department of Pharmacology.,Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany
| | - Nina Wettschureck
- Department of Pharmacology.,Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany
| |
Collapse
|
30
|
Aqmasheh S, Shamsasanjan K, Akbarzadehlaleh P, Pashoutan Sarvar D, Timari H. Effects of Mesenchymal Stem Cell Derivatives on Hematopoiesis and Hematopoietic Stem Cells. Adv Pharm Bull 2017; 7:165-177. [PMID: 28761818 PMCID: PMC5527230 DOI: 10.15171/apb.2017.021] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 04/08/2017] [Accepted: 04/18/2017] [Indexed: 12/11/2022] Open
Abstract
Hematopoiesis is a balance among quiescence, self-renewal, proliferation, and differentiation, which is believed to be firmly adjusted through interactions between hematopoietic stem and progenitor cells (HSPCs) with the microenvironment. This microenvironment is derived from a common progenitor of mesenchymal origin and its signals should be capable of regulating the cellular memory of transcriptional situation and lead to an exchange of stem cell genes expression. Mesenchymal stem cells (MSCs) have self-renewal and differentiation capacity into tissues of mesodermal origin, and these cells can support hematopoiesis through release various molecules that play a crucial role in migration, homing, self-renewal, proliferation, and differentiation of HSPCs. Studies on the effects of MSCs on HSPC differentiation can develop modern solutions in the treatment of patients with hematologic disorders for more effective Bone Marrow (BM) transplantation in the near future. However, considerable challenges remain on realization of how paracrine mechanisms of MSCs act on the target tissues, and how to design a therapeutic regimen with various paracrine factors in order to achieve optimal results for tissue conservation and regeneration. The aim of this review is to characterize and consider the related aspects of the ability of MSCs secretome in protection of hematopoiesis.
Collapse
Affiliation(s)
- Sara Aqmasheh
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Karim Shamsasanjan
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parvin Akbarzadehlaleh
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Hamze Timari
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
31
|
Heinrich V, Simpson WD, Francis EA. Analytical Prediction of the Spatiotemporal Distribution of Chemoattractants around Their Source: Theory and Application to Complement-Mediated Chemotaxis. Front Immunol 2017; 8:578. [PMID: 28603522 PMCID: PMC5445147 DOI: 10.3389/fimmu.2017.00578] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 05/01/2017] [Indexed: 11/13/2022] Open
Abstract
The ability of motile immune cells to detect and follow gradients of chemoattractant is critical to numerous vital functions, including their recruitment to sites of infection and-in emerging immunotherapeutic applications-to malignant tumors. Facilitated by a multitude of chemotactic receptors, the cells navigate a maze of stimuli to home in on their target. Distinct chemotactic processes direct this navigation at particular times and cell-target distances. The expedient coordination of this spatiotemporal hierarchy of chemotactic stages is the central element of a key paradigm of immunotaxis. Understanding this hierarchy is an enormous interdisciplinary challenge that requires, among others, quantitative insight into the shape, range, and dynamics of the profiles of chemoattractants around their sources. We here present a closed-form solution to a diffusion-reaction problem that describes the evolution of the concentration gradient of chemoattractant under various conditions. Our ready-to-use mathematical prescription captures many biological situations reasonably well and can be explored with standard graphing software, making it a valuable resource for every researcher studying chemotaxis. We here apply this mathematical model to characterize the chemoattractant cloud of anaphylatoxins that forms around bacterial and fungal pathogens in the presence of host serum. We analyze the spatial reach, rate of formation, and rate of dispersal of this locator cloud under realistic physiological conditions. Our analysis predicts that simply being small is an effective protective strategy of pathogens against complement-mediated discovery by host immune cells over moderate-to-large distances. Leveraging our predictions against single-cell, pure-chemotaxis experiments that use human immune cells as biosensors, we are able to explain the limited distance over which the cells recognize microbes. We conclude that complement-mediated chemotaxis is a universal, but short-range, homing mechanism by which chemotaxing immune cells can implement a last-minute course correction toward pathogenic microbes. Thus, the integration of theory and experiments provides a sound mechanistic explanation of the primary role of complement-mediated chemotaxis within the hierarchy of immunotaxis, and why other chemotactic processes are required for the successful recruitment of immune cells over large distances.
Collapse
Affiliation(s)
- Volkmar Heinrich
- Department of Biomedical Engineering, University of California at Davis, Davis, CA, United States
| | - Wooten D Simpson
- Department of Biomedical Engineering, University of California at Davis, Davis, CA, United States
| | - Emmet A Francis
- Department of Biomedical Engineering, University of California at Davis, Davis, CA, United States
| |
Collapse
|
32
|
Villarreal CDV, Alanis JCS, Pérez JCJ, Candiani JO. Cutaneous graft-versus-host disease after hematopoietic stem cell transplant - a review. An Bras Dermatol 2017; 91:336-43. [PMID: 27438202 PMCID: PMC4938279 DOI: 10.1590/abd1806-4841.20164180] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 01/27/2015] [Indexed: 11/22/2022] Open
Abstract
Graft-versus-host disease (GVHD) is a major complication of allogeneic
hematopoietic stem cell transplants (allo-HSCT) associated with significant
morbidity and mortality. The earliest and most common manifestation is cutaneous
graft-versus-host disease. This review focuses on the pathophysiology, clinical
features, prevention and treatment of cutaneous graft-versus-host disease. We
discuss various insights into the disease's mechanisms and the different
treatments for acute and chronic skin graft-versus-host disease.
Collapse
Affiliation(s)
| | | | - Jose Carlos Jaime Pérez
- "Dr. José E. Gonzalez" University Hospital of the ''Universidad Autonoma de Nuevo León'' Medical School - Nuevo León, Mexico
| | - Jorge Ocampo Candiani
- "Dr. José E. Gonzalez" University Hospital of the ''Universidad Autonoma de Nuevo León'' Medical School - Nuevo León, Mexico
| |
Collapse
|
33
|
T Lymphocytes and Inflammatory Mediators in the Interplay between Brain and Blood in Alzheimer's Disease: Potential Pools of New Biomarkers. J Immunol Res 2017; 2017:4626540. [PMID: 28293644 PMCID: PMC5331319 DOI: 10.1155/2017/4626540] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 12/22/2016] [Accepted: 01/23/2017] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disorder and the main cause of dementia. The disease is among the leading medical concerns of the modern world, because only symptomatic therapies are available, and no reliable, easily accessible biomarkers exist for AD detection and monitoring. Therefore extensive research is conducted to elucidate the mechanisms of AD pathogenesis, which seems to be heterogeneous and multifactorial. Recently much attention has been given to the neuroinflammation and activation of glial cells in the AD brain. Reports also highlighted the proinflammatory role of T lymphocytes infiltrating the AD brain. However, in AD molecular and cellular alterations involving T cells and immune mediators occur not only in the brain, but also in the blood and the cerebrospinal fluid (CSF). Here we review alterations concerning T lymphocytes and related immune mediators in the AD brain, CSF, and blood and the mechanisms by which peripheral T cells cross the blood brain barrier and the blood-CSF barrier. This knowledge is relevant for better AD therapies and for identification of novel biomarkers for improved AD diagnostics in the blood and the CSF. The data will be reviewed with the special emphasis on possibilities for development of AD biomarkers.
Collapse
|
34
|
A Novel Rabies Vaccine Expressing CXCL13 Enhances Humoral Immunity by Recruiting both T Follicular Helper and Germinal Center B Cells. J Virol 2017; 91:JVI.01956-16. [PMID: 27852854 DOI: 10.1128/jvi.01956-16] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 11/09/2016] [Indexed: 12/24/2022] Open
Abstract
Rabies remains a public health threat in most parts of the world, and approximately 99% of the cases are transmitted by dogs. There is an urgent need to develop an efficacious and affordable vaccine to control canine-transmitted rabies in developing countries. Our previous studies demonstrate that overexpression of chemokines/cytokines such as CCL-3 (MIP-1α) and granulocyte-macrophage colony-stimulating factor (GM-CSF) can enhance the immunogenicity of rabies vaccines. In the present study, the chemokine CXCL13 was inserted into the genome of the recombinant rabies virus (rRABV) strain LBNSE, and the effect of the chemokine CXCL13 on the immunogenicity of RABV was investigated. It was found that LBNSE-CXCL13 recruited follicular helper T (Tfh) and germinal center (GC) B cells, promoted the formation of GCs, and increased the population of plasma cells in immunized mice. Further studies showed that mice immunized with LBNSE-CXCL13 produced more rabies virus-neutralizing antibodies (VNAs) and developed better protection than those immunized with the parent virus LBNSE or the GM-CSF-expressing RABV (LBNSE-GM-CSF). Collectively, these findings provide a better understanding of the role of CXCL13 expression in the immunogenicity of the RABV, which may help in designing more-efficacious rabies vaccines. IMPORTANCE Rabies is endemic in most parts of the world, and more effort is needed to develop affordable and effective vaccines to control or eliminate this disease. The chemokine CXCL13 recruits both Tfh and B cells, which is essential for the homing of Tfh cells and the development of B cell follicles. In this study, the effect of the overexpression of CXCL13 on the immunogenicity of the RABV was evaluated in a mouse model. We found that CXCL13 expression promoted humoral immunity by recruiting Tfh and GC B cells, facilitating the formation of GCs, and increasing the number of plasma cells. As expected, the overexpression of CXCL13 resulted in enhanced virus-neutralizing antibody (VNA) production and protection against a virulent RABV challenge. These findings provide a better understanding of the role of CXCL13 in RABV-induced immune responses, which will help in designing more efficacious rabies vaccines.
Collapse
|
35
|
Development of T follicular helper cells and their role in disease and immune system. Biomed Pharmacother 2016; 84:1668-1678. [DOI: 10.1016/j.biopha.2016.10.083] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 10/26/2016] [Accepted: 10/26/2016] [Indexed: 02/06/2023] Open
|
36
|
Sepahi A, Casadei E, Tacchi L, Muñoz P, LaPatra SE, Salinas I. Tissue Microenvironments in the Nasal Epithelium of Rainbow Trout (Oncorhynchus mykiss) Define Two Distinct CD8α+ Cell Populations and Establish Regional Immunity. THE JOURNAL OF IMMUNOLOGY 2016; 197:4453-4463. [PMID: 27798156 DOI: 10.4049/jimmunol.1600678] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 09/29/2016] [Indexed: 12/17/2022]
Abstract
Mucosal surfaces require balancing different physiological roles and immune functions. To effectively achieve multifunctionality, mucosal epithelia have evolved unique microenvironments that create unique regional immune responses without impairing other normal physiological functions. Whereas examples of regional immunity are known in other mucosal epithelia, to date, no immune microenvironments have been described in the nasal mucosa, a site where the complex functions of olfaction and immunity need to be orchestrated. In this study we identified the presence of CD8α+ cells in the rainbow trout (Oncorhynchus mykiss) nasal epithelium. Nasal CD8α+ cells display a distinct phenotype suggestive of CD8+ T cells with high integrin β2 expression. Importantly, nasal CD8α+ cells are located in clusters at the mucosal tip of each olfactory lamella but scattered in the neuroepithelial region. The grouping of CD8α+ cells may be explained by the greater expression of CCL19, ICAM-1, and VCAM-1 in the mucosal tip compared with the neuroepithelium. Whereas viral Ag uptake occurred via both tip and lateral routes, tip-resident MHC class II+ cells are located significantly closer to the lumen of the nasal cavity than are their neuroepithelial counterparts, therefore having quicker access to invading pathogens. Our studies reveal compartmentalized mucosal immune responses within the nasal mucosa of a vertebrate species, a strategy that likely optimizes local immune responses while protecting olfactory sensory functions.
Collapse
Affiliation(s)
- Ali Sepahi
- Center for Evolutionary and Theoretical Immunology, Department of Biology, University of New Mexico, Albuquerque, NM 87131
| | - Elisa Casadei
- Center for Evolutionary and Theoretical Immunology, Department of Biology, University of New Mexico, Albuquerque, NM 87131
| | - Luca Tacchi
- Center for Evolutionary and Theoretical Immunology, Department of Biology, University of New Mexico, Albuquerque, NM 87131
| | - Pilar Muñoz
- Departamento de Sanidad Animal, Facultad de Veterinaria, Campus de Excelencia Internacional Regional Campus Mare Nostrum, Universidad de Murcia, 30100 Murcia, Spain; and
| | | | - Irene Salinas
- Center for Evolutionary and Theoretical Immunology, Department of Biology, University of New Mexico, Albuquerque, NM 87131;
| |
Collapse
|
37
|
Abstract
The chemokine, C-X-C motif ligand 13 (CXCL13), is constitutively expressed in lymphoid organs and controls the recruitment and compartmentalization of lymphocytes and antigen presenting cells within these specialized structures. Recent data, however, also show induction of this molecule under a variety of circumstances during central nervous system (CNS) inflammation. While its role(s) in the pathogenesis of neoplastic, infectious and autoimmune disorders of the CNS remain incompletely understood, growing evidence suggests that CXCL13 could become a relevant therapeutic target in at least some of these conditions. This review focuses on the diseases, cellular sources and external factors known to regulate CXCL13 production in the inflamed CNS.
Collapse
Affiliation(s)
- David N Irani
- Department of Neurology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
38
|
Haubruck P, Kammerer A, Korff S, Apitz P, Xiao K, Büchler A, Biglari B, Zimmermann G, Daniel V, Schmidmaier G, Moghaddam A. The treatment of nonunions with application of BMP-7 increases the expression pattern for angiogenic and inflammable cytokines: a matched pair analysis. J Inflamm Res 2016; 9:155-165. [PMID: 27703392 PMCID: PMC5036623 DOI: 10.2147/jir.s110621] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The local application of bone morphogenetic protein-7 (BMP-7) in combination with the transplantation of autologous bone graft improves the outcome in nonunion treatment; however, the specific reasons remain unclear. In this study, we sought to determine if the local application of BMP-7 contributes to improved bone regeneration in nonunion therapy by modulation of the angiogenic and inflammable cytokine expression patterns of the early inflammation response. Therefore, we utilized the analysis of serological cytokine expression patterns. As a matched pair analysis, best-fitting patients who were treated with transplantation of autologous bone graft (G1, n=10) were compared with patients who were treated with additional application of BMP-7 (G2, n=10). The changes in the cytokine expression patterns were monitored and correlated to clinical data of bone healing. Significant differences in angiogenesis potential (vascular endothelial growth factor [VEGF] serum levels) could be found in the first days after surgery (P<0.05). Furthermore, the increase and absolute amount of VEGF levels in the BMP-7 group were considerably higher than in the control group during the first 2 weeks after surgery. The expression pattern of inflammable cytokines showed noticeable differences in the time point of significant elevated levels, in particular, inflammable cytokines showed an earlier peak in G2. Furthermore, interleukin-6 was significantly elevated within the first week only, comparing G2 to G1 (P<0.05). Our findings indicate that BMP-7 induces an early and more intense expression of VEGF via a direct and postulated indirect pathway, thereby providing a favorable environment for bone healing. Moreover, application of BMP-7 leads to an earlier expression of known proinflammatory cytokines. The results of this study show that application of BMP-7 leads to costimulatory effect on both angiogenic and inflammable cytokine expression patterns that may serve as a possible stimulus for bone regeneration.
Collapse
Affiliation(s)
- Patrick Haubruck
- Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, Trauma and Reconstructive Surgery, Heidelberg University Hospital, HTRG - Heidelberg Trauma Research Group, Heidelberg, Germany
| | - Andreas Kammerer
- Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, Trauma and Reconstructive Surgery, Heidelberg University Hospital, HTRG - Heidelberg Trauma Research Group, Heidelberg, Germany
| | - Sebastian Korff
- Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, Trauma and Reconstructive Surgery, Heidelberg University Hospital, HTRG - Heidelberg Trauma Research Group, Heidelberg, Germany
| | - Philipp Apitz
- Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, Trauma and Reconstructive Surgery, Heidelberg University Hospital, HTRG - Heidelberg Trauma Research Group, Heidelberg, Germany
| | - Kai Xiao
- Department of Orthopedics Wuhan, Wuhan Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Axel Büchler
- Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, Trauma and Reconstructive Surgery, Heidelberg University Hospital, HTRG - Heidelberg Trauma Research Group, Heidelberg, Germany
| | - Bahram Biglari
- Department of Paraplegiology, Berufsgenossenschaftliche Unfallklinik Ludwigshafen, Ludwigshafen
| | - Gerald Zimmermann
- Department for Trauma Surgery, Theresienkrankenhaus und St. Hedwigs-Klinik GmbH, Mannheim
| | - Volker Daniel
- Department of Transplantation Immunology, Institute of Immunology, University of Heidelberg, Heidelberg, Germany
| | - Gerhard Schmidmaier
- Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, Trauma and Reconstructive Surgery, Heidelberg University Hospital, HTRG - Heidelberg Trauma Research Group, Heidelberg, Germany
| | - Arash Moghaddam
- Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, Trauma and Reconstructive Surgery, Heidelberg University Hospital, HTRG - Heidelberg Trauma Research Group, Heidelberg, Germany
| |
Collapse
|
39
|
Pals ST, Kersten MJ, Spaargaren M. Targeting cell adhesion and homing as strategy to cure Waldenström's macroglobulinemia. Best Pract Res Clin Haematol 2016; 29:161-168. [PMID: 27825462 DOI: 10.1016/j.beha.2016.08.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 08/29/2016] [Accepted: 08/30/2016] [Indexed: 12/19/2022]
Abstract
Most B-cell malignancies strictly depend on signals from the microenvironment for their survival and proliferation. This niche-dependency can be regarded as their Achilles' heel and provides an excellent target for therapy. Waldenström's macroglobulinemia (WM) is characterized by the accumulation of neoplastic post-germinal center B cells within the bone marrow (BM). Interestingly, one third of the patients carry activating mutations in the chemokine receptor CXCR4, a key mediator of B cell and plasma cell homing to the BM. We have previously shown that signals from the B-cell antigen receptor (BCR) and from chemokine receptors play a central role in controlling the interaction of normal and malignant B cells with their microenvironment by regulating the activity of integrin adhesion molecules. Apart from controlling the homing and retention of lymphocytes within their growth- and survival niches, integrins also emit signals that directly promote cell growth and survival. By analogy to the successful treatment with BTK inhibitors, we propose that targeting pathways controlling integrin-mediated retention of the WM cells in the BM, thereby inducing 'homelessness' (anoikis) by mobilization of the malignant cells from their protective niches, may be an efficient treatment strategy for WM.
Collapse
Affiliation(s)
- Steven T Pals
- Department of Pathology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; Lymphoma and Myeloma Center Amsterdam - LYMMCARE, The Netherlands.
| | - Marie José Kersten
- Department of Hematology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; Lymphoma and Myeloma Center Amsterdam - LYMMCARE, The Netherlands
| | - Marcel Spaargaren
- Department of Pathology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; Lymphoma and Myeloma Center Amsterdam - LYMMCARE, The Netherlands
| |
Collapse
|
40
|
Herrmann MM, Barth S, Greve B, Schumann KM, Bartels A, Weissert R. Identification of gene expression patterns crucially involved in experimental autoimmune encephalomyelitis and multiple sclerosis. Dis Model Mech 2016; 9:1211-1220. [PMID: 27519689 PMCID: PMC5087830 DOI: 10.1242/dmm.025536] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 08/09/2016] [Indexed: 01/22/2023] Open
Abstract
After encounter with a central nervous system (CNS)-derived autoantigen, lymphocytes leave the lymph nodes and enter the CNS. This event leads only rarely to subsequent tissue damage. Genes relevant to CNS pathology after cell infiltration are largely undefined. Myelin-oligodendrocyte-glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE) is an animal model of multiple sclerosis (MS), a chronic autoimmune disease of the CNS that results in disability. To assess genes that are involved in encephalitogenicity and subsequent tissue damage mediated by CNS-infiltrating cells, we performed a DNA microarray analysis from cells derived from lymph nodes and eluted from CNS in LEW.1AV1 (RT1av1) rats immunized with MOG 91-108. The data was compared to immunizations with adjuvant alone or naive rats and to immunizations with the immunogenic but not encephalitogenic MOG 73-90 peptide. Here, we show involvement of Cd38, Cxcr4 and Akt and confirm these findings by the use of Cd38-knockout (B6.129P2-Cd38tm1Lnd/J) mice, S1P-receptor modulation during EAE and quantitative expression analysis in individuals with MS. The hereby-defined underlying pathways indicate cellular activation and migration pathways mediated by G-protein-coupled receptors as crucial events in CNS tissue damage. These pathways can be further explored for novel therapeutic interventions. Summary: To define disease-inducing immune responses, unbiased gene expression in LEW.1AV1 (RT1av1) rats immunized with autoantigen-derived encephalitogenic and non-encephalitogenic peptides was investigated in MOG-EAE. CXCR4, CXCL12, AKT and CD38 control CNS inflammation.
Collapse
Affiliation(s)
- Martin M Herrmann
- Department of General Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany
| | - Silvia Barth
- Department of General Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany
| | - Bernhard Greve
- Department of General Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany
| | - Kathrin M Schumann
- Department of General Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany
| | - Andrea Bartels
- Department of General Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany
| | - Robert Weissert
- Department of General Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany Department of Neurology, University of Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
41
|
Liu X, Asokan SB, Bear JE, Haugh JM. Quantitative analysis of B-lymphocyte migration directed by CXCL13. Integr Biol (Camb) 2016; 8:894-903. [PMID: 27477203 DOI: 10.1039/c6ib00128a] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
B-lymphocyte migration, directed by chemokine gradients, is essential for homing to sites of antigen presentation. B cells move rapidly, exhibiting amoeboid morphology like other leukocytes, yet quantitative studies addressing B-cell migration are currently lacking relative to neutrophils, macrophages, and T cells. Here, we used total internal reflection fluorescence (TIRF) microscopy to characterize the changes in shape (morphodynamics) of primary, murine B cells as they migrated on surfaces with adsorbed chemokine, CXCL13, and the adhesive ligand, ICAM-1. B cells exhibited frequent, spontaneous dilation and shrinking events at the sides of the leading membrane edge, a phenomenon that was predictive of turning versus directional persistence. To characterize directed B-cell migration, a microfluidic device was implemented to generate gradients of adsorbed CXCL13 gradients. Haptotaxis assays revealed a modest yet consistently positive bias of the cell's persistent random walk behavior towards CXCL13 gradients. Quantification of tactic fidelity showed that bias is optimized by steeper gradients without excessive midpoint density of adsorbed chemokine. Under these conditions, B-cell migration is more persistent when the direction of migration is better aligned with the gradient.
Collapse
Affiliation(s)
- Xiaji Liu
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Campus Box 7905, 911 Partners Way, Raleigh, NC 27695-7905, USA.
| | | | | | | |
Collapse
|
42
|
Fu H, Ward EJ, Marelli-Berg FM. Mechanisms of T cell organotropism. Cell Mol Life Sci 2016; 73:3009-33. [PMID: 27038487 PMCID: PMC4951510 DOI: 10.1007/s00018-016-2211-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 03/21/2016] [Accepted: 03/22/2016] [Indexed: 02/06/2023]
Abstract
Protective immunity relies upon T cell differentiation and subsequent migration to target tissues. Similarly, immune homeostasis requires the localization of regulatory T cells (Tregs) to the sites where immunity takes place. While naïve T lymphocytes recirculate predominantly in secondary lymphoid tissue, primed T cells and activated Tregs must traffic to the antigen rich non-lymphoid tissue to exert effector and regulatory responses, respectively. Following priming in draining lymph nodes, T cells acquire the 'homing receptors' to facilitate their access to specific tissues and organs. An additional level of topographic specificity is provided by T cells receptor recognition of antigen displayed by the endothelium. Furthermore, co-stimulatory signals (such as those induced by CD28) have been shown not only to regulate T cell activation and differentiation, but also to orchestrate the anatomy of the ensuing T cell response. We here review the molecular mechanisms supporting trafficking of both effector and regulatory T cells to specific antigen-rich tissues.
Collapse
Affiliation(s)
- Hongmei Fu
- William Harvey Research Institute, Heart Centre, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Eleanor Jayne Ward
- William Harvey Research Institute, Heart Centre, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Federica M Marelli-Berg
- William Harvey Research Institute, Heart Centre, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK.
| |
Collapse
|
43
|
Expression of the chemokine receptor gene, CCR8, is associated With DUSP22 rearrangements in anaplastic large cell lymphoma. Appl Immunohistochem Mol Morphol 2016; 23:580-9. [PMID: 25390351 DOI: 10.1097/pai.0000000000000118] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Anaplastic large cell lymphoma (ALCL) is one of the most common T-cell non-Hodgkin lymphomas and has 2 main subtypes: an anaplastic lymphoma kinase (ALK)-positive subtype characterized by ALK gene rearrangements and an ALK-negative subtype that is poorly understood. We recently identified recurrent rearrangements of the DUSP22 locus on 6p25.3 in both primary cutaneous and systemic ALK-negative ALCLs. This study aimed to determine the relationship between these rearrangements and expression of the chemokine receptor gene, CCR8. CCR8 has skin-homing properties and has been suggested to play a role in limiting extracutaneous spread of primary cutaneous ALCLs. However, overexpression of CCR8 has also been reported in systemic ALK-negative ALCLs. As available antibodies for CCR8 have shown lack of specificity, we examined CCR8 expression using quantitative real-time PCR in frozen tissue and RNA in situ hybridization (ISH) in paraffin tissue. Both approaches showed higher CCR8 expression in ALCLs with DUSP22 rearrangements than in nonrearranged cases (PCR: 19.5-fold increase, P=0.01; ISH: 3.3-fold increase, P=0.0008). CCR8 expression was not associated with cutaneous presentation, cutaneous biopsy site, or cutaneous involvement during the disease course. These findings suggest that CCR8 expression in ALCL is more closely related to the presence of DUSP22 rearrangements than to cutaneous involvement and that the function of CCR8 may extend beyond its skin-homing properties in this disease. This study also underscores the utility of RNA-ISH as a paraffin-based method for investigating gene expression when reliable antibodies for immunohistochemical analysis are not available.
Collapse
|
44
|
Amer LD, Bryant SJ. The In Vitro and In Vivo Response to MMP-Sensitive Poly(Ethylene Glycol) Hydrogels. Ann Biomed Eng 2016; 44:1959-69. [PMID: 27080375 PMCID: PMC5577801 DOI: 10.1007/s10439-016-1608-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 04/02/2016] [Indexed: 12/28/2022]
Abstract
Enzyme-sensitive hydrogels are a promising class of materials for cell encapsulation and tissue engineering because their ability to be degraded by cell-secreted factors. However, it is well known that nearly all synthetic biomaterials elicit a foreign body response (FBR) upon implantation. Therefore, this study aimed to evaluate the in vitro and in vivo response to an enzyme-sensitive hydrogel. Hydrogels were formed from poly(ethylene glycol) with the peptide crosslinker, C-VPLS↓LYSG-C, which is susceptible to matrix metalloproteinases 2 and 9. We evaluated the hydrogel by exogenously delivered enzymes, encapsulated mesenchymal stem cells as a tissue engineering relevant cell type, and by macrophage-secreted factors in vitro and for the FBR through macrophage attachment in vitro and in a subcutaneous mouse model. These hydrogels rapidly degraded upon exposure to exogenous MMP-2 and to lesser degree with MMP-9. Encapsulated mesenchymal stem cells were capable of degrading the hydrogels via matrix metalloproteinases. Inflammatory macrophages were confirmed to attach to the hydrogels, but were not capable of rapidly degrading the hydrogels. In vivo, these hydrogels remained intact after 4 weeks and exhibited a classic FBR with inflammatory cells at the hydrogel surface and a fibrous capsule. In summary, these findings suggest that while this MMP-2/9 sensitive hydrogel is readily degraded in vitro, it does not undergo rapid degradation by the FBR. Thus, the long term stability of these hydrogels in vivo coupled with the ability for encapsulated cells to degrade the hydrogel makes them promising materials for tissue engineering.
Collapse
Affiliation(s)
- Luke D Amer
- Department of Chemical and Biological Engineering, University of Colorado, 3415 Colorado Ave, UCB 596, Boulder, CO, 80303, USA
- BioFrontiers Institute, University of Colorado, 3415 Colorado Avenue, Boulder, CO, 80303, USA
| | - Stephanie J Bryant
- Department of Chemical and Biological Engineering, University of Colorado, 3415 Colorado Ave, UCB 596, Boulder, CO, 80303, USA.
- BioFrontiers Institute, University of Colorado, 3415 Colorado Avenue, Boulder, CO, 80303, USA.
- Material Science and Engineering Program, University of Colorado, 3415 Colorado Avenue, Boulder, CO, 80303, USA.
| |
Collapse
|
45
|
Azzi J, Yin Q, Uehara M, Ohori S, Tang L, Cai K, Ichimura T, McGrath M, Maarouf O, Kefaloyianni E, Loughhead S, Petr J, Sun Q, Kwon M, Tullius S, von Andrian UH, Cheng J, Abdi R. Targeted Delivery of Immunomodulators to Lymph Nodes. Cell Rep 2016; 15:1202-13. [PMID: 27134176 PMCID: PMC4973867 DOI: 10.1016/j.celrep.2016.04.007] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 02/21/2016] [Accepted: 03/28/2016] [Indexed: 11/03/2022] Open
Abstract
Active-targeted delivery to lymph nodes represents a major advance toward more effective treatment of immune-mediated disease. The MECA79 antibody recognizes peripheral node addressin molecules expressed by high endothelial venules of lymph nodes. By mimicking lymphocyte trafficking to the lymph nodes, we have engineered MECA79-coated microparticles containing an immunosuppressive medication, tacrolimus. Following intravenous administration, MECA79-bearing particles showed marked accumulation in the draining lymph nodes of transplanted animals. Using an allograft heart transplant model, we show that targeted lymph node delivery of microparticles containing tacrolimus can prolong heart allograft survival with negligible changes in tacrolimus serum level. Using MECA79 conjugation, we have demonstrated targeted delivery of tacrolimus to the lymph nodes following systemic administration, with the capacity for immune modulation in vivo.
Collapse
Affiliation(s)
- Jamil Azzi
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Qian Yin
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61820, USA
| | - Mayuko Uehara
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Shunsuke Ohori
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Li Tang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61820, USA
| | - Kaimin Cai
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61820, USA
| | - Takaharu Ichimura
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Martina McGrath
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Omar Maarouf
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Eirini Kefaloyianni
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Scott Loughhead
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Jarolim Petr
- Department of Pathology, Clinical Laboratories Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Qidi Sun
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61820, USA
| | - Mincheol Kwon
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61820, USA
| | - Stefan Tullius
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ulrich H von Andrian
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA; The Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Jianjun Cheng
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61820, USA.
| | - Reza Abdi
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
46
|
Abstract
Bacillus anthracis is killed by the interferon-inducible, ELR(−) CXC chemokine CXCL10. Previous studies showed that disruption of the gene encoding FtsX, a conserved membrane component of the ATP-binding cassette transporter-like complex FtsE/X, resulted in resistance to CXCL10. FtsX exhibits some sequence similarity to the mammalian CXCL10 receptor, CXCR3, suggesting that the CXCL10 N-terminal region that interacts with CXCR3 may also interact with FtsX. A C-terminal truncated CXCL10 was tested to determine if the FtsX-dependent antimicrobial activity is associated with the CXCR3-interacting N terminus. The truncated CXCL10 exhibited antimicrobial activity against the B. anthracis parent strain but not the ΔftsX mutant, which supports a key role for the CXCL10 N terminus. Mutations in FtsE, the conserved ATP-binding protein of the FtsE/X complex, resulted in resistance to both CXCL10 and truncated CXCL10, indicating that both FtsX and FtsE are important. Higher concentrations of CXCL10 overcame the resistance of the ΔftsX mutant to CXCL10, suggesting an FtsX-independent killing mechanism, likely involving its C-terminal α-helix, which resembles a cationic antimicrobial peptide. Membrane depolarization studies revealed that CXCL10 disrupted membranes of the B. anthracis parent strain and the ΔftsX mutant, but only the parent strain underwent depolarization with truncated CXCL10. These findings suggest that CXCL10 is a bifunctional molecule that kills B. anthracis by two mechanisms. FtsE/X-dependent killing is mediated through an N-terminal portion of CXCL10 and is not reliant upon the C-terminal α-helix. The FtsE/X-independent mechanism involves membrane depolarization by CXCL10, likely because of its α-helix. These findings present a new paradigm for understanding mechanisms by which CXCL10 and related chemokines kill bacteria. Chemokines are a class of molecules known for their chemoattractant properties but more recently have been shown to possess antimicrobial activity against a wide range of Gram-positive and Gram-negative bacterial pathogens. The mechanism(s) by which these chemokines kill bacteria is not well understood, but it is generally thought to be due to the conserved amphipathic C-terminal α-helix that resembles cationic antimicrobial peptides in charge and secondary structure. Our present study indicates that the interferon-inducible, ELR(−) chemokine CXCL10 kills the Gram-positive pathogen Bacillus anthracis through multiple molecular mechanisms. One mechanism is mediated by interaction of CXCL10 with the bacterial FtsE/X complex and does not require the presence of the CXCL10 C-terminal α-helix. The second mechanism is FtsE/X receptor independent and kills through membrane disruption due to the C-terminal α-helix. This study represents a new paradigm for understanding how chemokines exert an antimicrobial effect that may prove applicable to other bacterial species.
Collapse
|
47
|
Zirafi O, Hermann PC, Münch J. Proteolytic processing of human serum albumin generates EPI-X4, an endogenous antagonist of CXCR4. J Leukoc Biol 2016; 99:863-8. [PMID: 26965637 DOI: 10.1189/jlb.2mr1115-521rr] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 02/11/2016] [Indexed: 12/14/2022] Open
Abstract
The chemokine receptor CXCR4 is an important G protein-coupled receptor. Signaling via CXCL12 regulates a number of important biologic processes, including immune responses, organogenesis, or hematopoiesis. Dysregulation of CXCR4 signaling is associated with a variety of diseases, such as cancer development and metastasis, immunodeficiencies, or chronic inflammation. Here, we review our findings on endogenous peptide inhibitor of CXCR4 as a novel antagonist of CXCR4. This peptide is a 16-residue fragment of human serum albumin and was isolated as an inhibitor of CXCR4-tropic human immunodeficiency virus type 1 from a blood-derived peptide library. Endogenous peptide inhibitor of CXCR4 binds the second extracellular loop of CXCR4, thereby preventing engagement of CXCL12 and antagonizing the receptor. Consequently, endogenous peptide inhibitor of CXCR4 inhibits CXCL12-mediated migration of CXCR4-expressing cells in vitro, mobilizes hematopoietic stem cells, and suppresses inflammatory responses in vivo. We discuss the generation of endogenous peptide inhibitor of CXCR4, its relevance as biomarker for disease, and its role in human immunodeficiency virus/acquired immunodeficiency syndrome pathogenesis and cancer. Furthermore, we discuss why optimized endogenous peptide inhibitor of CXCR4 derivatives might have advantages over other CXCR4 antagonists.
Collapse
Affiliation(s)
- Onofrio Zirafi
- Institute of Molecular Virology, University of Ulm, Ulm, Germany
| | - Patrick C Hermann
- Department of Internal Medicine I, University of Ulm, Ulm, Germany; and
| | - Jan Münch
- Institute of Molecular Virology, University of Ulm, Ulm, Germany; Ulm Peptide Pharmaceuticals, University of Ulm, Ulm, Germany
| |
Collapse
|
48
|
Liu X, Welf ES, Haugh JM. Linking morphodynamics and directional persistence of T lymphocyte migration. J R Soc Interface 2016; 12:rsif.2014.1412. [PMID: 25904526 DOI: 10.1098/rsif.2014.1412] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
T cells play a central role in the adaptive immune response, and their directed migration is essential for homing to sites of antigen presentation. Like neutrophils, T lymphocytes are rapidly moving cells that exhibit amoeboid movement, characterized by a definitive polarity with F-actin concentrated at the front and myosin II elsewhere. In this study, we used total internal reflection fluorescence (TIRF) microscopy to monitor the cells' areas of contact with a surface presenting adhesive ICAM-1 and the chemokine, CXCL12/SDF-1. Our analysis reveals that T-cell migration and reorientation are achieved by bifurcation and lateral separation of protrusions along the leading membrane edge, followed by cessation of one of the protrusions, which acts as a pivot for cell turning. We show that the distribution of bifurcation frequencies exhibits characteristics of a random, spontaneous process; yet, the waiting time between bifurcation events depends on whether or not the pivot point remains on the same side of the migration axis. Our analysis further suggests that switching of the dominant protrusion between the two sides of the migration axis is associated with persistent migration, whereas the opposite is true of cell turning. To help explain the bifurcation phenomenon and how distinct migration behaviours might arise, a spatio-temporal, stochastic model describing F-actin dynamics is offered.
Collapse
Affiliation(s)
- Xiaji Liu
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Campus Box 7905, Raleigh, NC 27695, USA
| | - Erik S Welf
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Campus Box 7905, Raleigh, NC 27695, USA
| | - Jason M Haugh
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Campus Box 7905, Raleigh, NC 27695, USA
| |
Collapse
|
49
|
Hauser MA, Legler DF. Common and biased signaling pathways of the chemokine receptor CCR7 elicited by its ligands CCL19 and CCL21 in leukocytes. J Leukoc Biol 2016; 99:869-82. [PMID: 26729814 DOI: 10.1189/jlb.2mr0815-380r] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 12/17/2015] [Indexed: 12/24/2022] Open
Abstract
Chemokines are pivotal regulators of cell migration during continuous immune surveillance, inflammation, homeostasis, and development. Chemokine binding to their 7-transmembrane domain, G-protein-coupled receptors causes conformational changes that elicit intracellular signaling pathways to acquire and maintain an asymmetric architectural organization and a polarized distribution of signaling molecules necessary for directional cell migration. Leukocytes rely on the interplay of chemokine-triggered migration modules to promote amoeboid-like locomotion. One of the most important chemokine receptors for adaptive immune cell migration is the CC-chemokine receptor CCR7. CCR7 and its ligands CCL19 and CCL21 control homing of T cells and dendritic cells to areas of the lymph nodes where T cell priming and the initiation of the adaptive immune response occur. Moreover, CCR7 signaling also contributes to T cell development in the thymus and to lymphorganogenesis. Although the CCR7-CCL19/CCL21 axis evolved to benefit the host, inappropriate regulation or use of these proteins can contribute or cause pathobiology of chronic inflammation, tumorigenesis, and metastasis, as well as autoimmune diseases. Therefore, it appears as the CCR7-CCL19/CCL21 axis is tightly regulated at numerous intersections. Here, we discuss the multiple regulatory mechanism of CCR7 signaling and its influence on CCR7 function. In particular, we focus on the functional diversity of the 2 CCR7 ligands, CCL19 and CCL21, as well as on their impact on biased signaling. The understanding of the molecular determinants of biased signaling and the multiple layers of CCR7 regulation holds the promise for potential future therapeutic intervention.
Collapse
Affiliation(s)
- Mark A Hauser
- Biotechnology Institute Thurgau at the University of Konstanz, Kreuzlingen, Switzerland
| | - Daniel F Legler
- Biotechnology Institute Thurgau at the University of Konstanz, Kreuzlingen, Switzerland
| |
Collapse
|
50
|
Kim CH, Hashimoto-Hill S, Kim M. Migration and Tissue Tropism of Innate Lymphoid Cells. Trends Immunol 2015; 37:68-79. [PMID: 26708278 DOI: 10.1016/j.it.2015.11.003] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 11/08/2015] [Accepted: 11/12/2015] [Indexed: 12/15/2022]
Abstract
Innate lymphoid cell (ILCs) subsets differentially populate various barrier and non-barrier tissues, where they play important roles in tissue homeostasis and tissue-specific responses to pathogen attack. Recent findings have provided insight into the molecular mechanisms that guide ILC migration into peripheral tissues, revealing common features among different ILC subsets as well as important distinctions. Recent studies have also highlighted the impact of tissue-specific cues on ILC migration, and the importance of the local immunological milieu. We review these findings here and discuss how the migratory patterns and tissue tropism of different ILC subsets relate to the development and differentiation of these cells, and to ILC-mediated tissue-specific regulation of innate and adaptive immune responses. In this context we outline open questions and important areas of future research.
Collapse
Affiliation(s)
- Chang H Kim
- Laboratory of Immunology and Hematopoiesis, Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA; Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA.
| | - Seika Hashimoto-Hill
- Laboratory of Immunology and Hematopoiesis, Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA
| | - Myunghoo Kim
- Laboratory of Immunology and Hematopoiesis, Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|