1
|
Casais-E-Silva LL, Cruz LF, Dos Reis VP, Paloschi MV, Teixeira C, Zuliani JP, da Silva Setubal S. Micrurus lemniscatus venom stimulates leukocyte functions in vivo. Arch Toxicol 2025; 99:1591-1603. [PMID: 39948207 DOI: 10.1007/s00204-025-03970-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 01/27/2025] [Indexed: 04/04/2025]
Abstract
While Micrurus venoms are primarily recognized for inducing neurotoxic effects, experimental findings have also documented additional manifestations such as local effects such as edema, myotoxicity, and inflammation. However, limited information is available regarding the impact of Micrurus venom on leukocyte functions. In this study, we investigated the in vivo effects of Micrurus lemniscatus venom (ML venom) on peritoneal leukocyte functions. Intraperitoneal (i.p.) injection of ML venom stimulated leukocyte migration, particularly at lower doses, with predominance of mononuclear cells. Both doses also triggered the release of cytokines (TNF-α, IL-1β, and IL-6) three hours after injection. Additionally, ML venom elicited the production of reactive oxygen species (ROS) and hydrogen peroxide (H2O2), as well as enhanced phagocytosis, along with the release of dsDNA and lipid droplets by these cells. This study represents the first demonstration of peritoneal leukocyte activation by Micrurus lemniscatus venom.
Collapse
Affiliation(s)
- Luciana Lyra Casais-E-Silva
- Laboratório de Neuroimunoendocrinologia e Toxinologia, Instituto de Ciências da Saúde, Universidade Federal da Bahia (UFBA), Salvador, BA, Brazil.
| | - Larissa Faustina Cruz
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz (FIOCRUZ) Rondônia, Porto Velho, RO, Brazil
| | - Valdison P Dos Reis
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz (FIOCRUZ) Rondônia, Porto Velho, RO, Brazil
| | - Mauro V Paloschi
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz (FIOCRUZ) Rondônia, Porto Velho, RO, Brazil
| | - Catarina Teixeira
- Laboratório de Farmacologia-Instituto Butantan, São Paulo, SP, Brazil.
| | - Juliana P Zuliani
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz (FIOCRUZ) Rondônia, Porto Velho, RO, Brazil
- Departamento de Medicina, Universidade Federal de Rondônia (UNIR), Porto Velho, RO, Brazil
| | - Sulamita da Silva Setubal
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz (FIOCRUZ) Rondônia, Porto Velho, RO, Brazil.
| |
Collapse
|
2
|
Duan M, Xu Y, Li Y, Feng H, Chen Y. Targeting brain-peripheral immune responses for secondary brain injury after ischemic and hemorrhagic stroke. J Neuroinflammation 2024; 21:102. [PMID: 38637850 PMCID: PMC11025216 DOI: 10.1186/s12974-024-03101-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 04/15/2024] [Indexed: 04/20/2024] Open
Abstract
The notion that the central nervous system is an immunologically immune-exempt organ has changed over the past two decades, with increasing evidence of strong links and interactions between the central nervous system and the peripheral immune system, both in the healthy state and after ischemic and hemorrhagic stroke. Although primary injury after stroke is certainly important, the limited therapeutic efficacy, poor neurological prognosis and high mortality have led researchers to realize that secondary injury and damage may also play important roles in influencing long-term neurological prognosis and mortality and that the neuroinflammatory process in secondary injury is one of the most important influences on disease progression. Here, we summarize the interactions of the central nervous system with the peripheral immune system after ischemic and hemorrhagic stroke, in particular, how the central nervous system activates and recruits peripheral immune components, and we review recent advances in corresponding therapeutic approaches and clinical studies, emphasizing the importance of the role of the peripheral immune system in ischemic and hemorrhagic stroke.
Collapse
Affiliation(s)
- Mingxu Duan
- Department of Neurosurgery, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Ya Xu
- Department of Neurosurgery, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yuanshu Li
- Department of Neurosurgery, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Hua Feng
- Department of Neurosurgery, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yujie Chen
- Department of Neurosurgery, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
3
|
Awamura T, Nakasone ES, Gangcuangco LM, Subia NT, Bali AJ, Chow DC, Shikuma CM, Park J. Platelet and HIV Interactions and Their Contribution to Non-AIDS Comorbidities. Biomolecules 2023; 13:1608. [PMID: 38002289 PMCID: PMC10669125 DOI: 10.3390/biom13111608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 10/29/2023] [Accepted: 10/31/2023] [Indexed: 11/26/2023] Open
Abstract
Platelets are anucleate cytoplasmic cell fragments that circulate in the blood, where they are involved in regulating hemostasis. Beyond their normal physiologic role, platelets have emerged as versatile effectors of immune response. During an infection, cell surface receptors enable platelets to recognize viruses, resulting in their activation. Activated platelets release biologically active molecules that further trigger host immune responses to protect the body against infection. Their impact on the immune response is also associated with the recruitment of circulating leukocytes to the site of infection. They can also aggregate with leukocytes, including lymphocytes, monocytes, and neutrophils, to immobilize pathogens and prevent viral dissemination. Despite their host protective role, platelets have also been shown to be associated with various pathophysiological processes. In this review, we will summarize platelet and HIV interactions during infection. We will also highlight and discuss platelet and platelet-derived mediators, how they interact with immune cells, and the multifaceted responsibilities of platelets in HIV infection. Furthermore, we will give an overview of non-AIDS comorbidities linked to platelet dysfunction and the impact of antiretroviral therapy on platelet function.
Collapse
Affiliation(s)
- Thomas Awamura
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (T.A.); (N.T.S.); (A.-J.B.)
| | - Elizabeth S. Nakasone
- University of Hawai‘i Cancer Center, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA;
- Department of Medicine, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA;
| | - Louie Mar Gangcuangco
- Hawai‘i Center for AIDS, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (L.M.G.); (C.M.S.)
| | - Natalie T. Subia
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (T.A.); (N.T.S.); (A.-J.B.)
| | - Aeron-Justin Bali
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (T.A.); (N.T.S.); (A.-J.B.)
| | - Dominic C. Chow
- Department of Medicine, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA;
- Hawai‘i Center for AIDS, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (L.M.G.); (C.M.S.)
| | - Cecilia M. Shikuma
- Hawai‘i Center for AIDS, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (L.M.G.); (C.M.S.)
| | - Juwon Park
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (T.A.); (N.T.S.); (A.-J.B.)
- Hawai‘i Center for AIDS, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (L.M.G.); (C.M.S.)
| |
Collapse
|
4
|
Tang Y, Yu Z, Lu X, Fan Q, Huang W. Overcoming Vascular Barriers to Improve the Theranostic Outcomes of Nanomedicines. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103148. [PMID: 35246962 PMCID: PMC9069202 DOI: 10.1002/advs.202103148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 02/16/2022] [Indexed: 05/04/2023]
Abstract
Nanotheranostics aims to utilize nanomaterials to prevent, diagnose, and treat diseases to improve the quality of patients' lives. Blood vessels are responsible to deliver nutrients and oxygen to the whole body, eliminate waste, and provide access for patrolling immune cells for healthy tissues. Meanwhile, they can also nourish disease tissues, spread disease factors or cells into other healthy tissues, and deliver nanotheranostic agents to cover all the regions of a disease tissue. Thus, blood vessels are the first and the most important barrier for highly efficient nanotheranostics. Here, the structure and function of blood vessels are explored and how these characteristics affect nanotheranostics is discussed. Moreover, new mechanisms and related strategies about overcoming vascular obstacles for improved nanotheranostic outcomes are critically summarized, and their merits and demerits of each strategy are analyzed. Moreover, the present challenges to completely exhibit the potential of overcoming vascular barriers to improve the theranostic outcomes of nanomedicines in life science are also discussed. Finally, the future perspective is further discussed.
Collapse
Affiliation(s)
- Yufu Tang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM)Nanjing Tech University30 South Puzhu RoadNanjing211800P. R. China
| | - Zhongzheng Yu
- School of Chemical and Biomedical EngineeringNanyang Technological UniversitySingapore637459Singapore
| | - Xiaomei Lu
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM)Nanjing Tech University30 South Puzhu RoadNanjing211800P. R. China
| | - Quli Fan
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for BiosensorsInstitute of Advanced Materials (IAM)Nanjing University of Posts and TelecommunicationsNanjing210023China
- Shaanxi Institute of Flexible Electronics (SIFE)Northwestern Polytechnical University (NPU)Xi'an710072China
| | - Wei Huang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM)Nanjing Tech University30 South Puzhu RoadNanjing211800P. R. China
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for BiosensorsInstitute of Advanced Materials (IAM)Nanjing University of Posts and TelecommunicationsNanjing210023China
- Shaanxi Institute of Flexible Electronics (SIFE)Northwestern Polytechnical University (NPU)Xi'an710072China
| |
Collapse
|
5
|
da Silva PR, do Espírito Santo RF, Melo CDO, Pachú Cavalcante FE, Costa TB, Barbosa YV, e Silva YMSDM, de Sousa NF, Villarreal CF, de Moura RO, dos Santos VL. The Compound (E)-2-Cyano- N,3-diphenylacrylamide (JMPR-01): A Potential Drug for Treatment of Inflammatory Diseases. Pharmaceutics 2022; 14:188. [PMID: 35057082 PMCID: PMC8777680 DOI: 10.3390/pharmaceutics14010188] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/07/2022] [Accepted: 01/10/2022] [Indexed: 02/04/2023] Open
Abstract
The compound (E)-2-cyano-N,3-diphenylacrylamide (JMPR-01) was structurally developed using bioisosteric modifications of a hybrid prototype as formed from fragments of indomethacin and paracetamol. Initially, in vitro assays were performed to determine cell viability (in macrophage cultures), and its ability to modulate the synthesis of nitrite and cytokines (IL-1β and TNFα) in non-cytotoxic concentrations. In vivo, anti-inflammatory activity was explored using the CFA-induced paw edema and zymosan-induced peritonitis models. To investigate possible molecular targets, molecular docking was performed with the following crystallographic structures: LT-A4-H, PDE4B, COX-2, 5-LOX, and iNOS. As results, we observed a significant reduction in the production of nitrite and IL-1β at all concentrations used, and also for TNFα with JMPR-01 at 50 and 25 μM. The anti-edematogenic activity of JMPR-01 (100 mg/kg) was significant, reducing edema at 2-6 h, similar to the dexamethasone control. In induced peritonitis, JMPR-01 reduced leukocyte migration by 61.8, 68.5, and 90.5% at respective doses of 5, 10, and 50 mg/kg. In silico, JMPR-01 presented satisfactory coupling; mainly with LT-A4-H, PDE4B, and iNOS. These preliminary results demonstrate the strong potential of JMPR-01 to become a drug for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Pablo Rayff da Silva
- Programa de Pós Graduação em Ciências Farmacêuticas, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil; (P.R.d.S.); camillamello-@hotmail.com (C.d.O.M.); (R.O.d.M.)
- Laboratório de Ensaios Farmacológicos, Departamento de Farmácia, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil; (F.E.P.C.); (T.B.C.); (Y.V.B.)
- Laboratório de Desenvolvimento e Síntese de Fármacos, Departamento de Farmácia, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil;
| | - Renan Fernandes do Espírito Santo
- Instituto Gonçalo Moniz, Fundação Osvaldo Cruz, Salvador 40296-710, BA, Brazil; (R.F.d.E.S.); (C.F.V.)
- Faculdade de Farmácia, Universidade Federal da Bahia, Salvador 40170-290, BA, Brazil
| | - Camila de Oliveira Melo
- Programa de Pós Graduação em Ciências Farmacêuticas, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil; (P.R.d.S.); camillamello-@hotmail.com (C.d.O.M.); (R.O.d.M.)
- Laboratório de Desenvolvimento e Síntese de Fármacos, Departamento de Farmácia, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil;
| | - Fábio Emanuel Pachú Cavalcante
- Laboratório de Ensaios Farmacológicos, Departamento de Farmácia, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil; (F.E.P.C.); (T.B.C.); (Y.V.B.)
| | - Thássia Borges Costa
- Laboratório de Ensaios Farmacológicos, Departamento de Farmácia, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil; (F.E.P.C.); (T.B.C.); (Y.V.B.)
| | - Yasmim Vilarim Barbosa
- Laboratório de Ensaios Farmacológicos, Departamento de Farmácia, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil; (F.E.P.C.); (T.B.C.); (Y.V.B.)
| | - Yvnni M. S. de Medeiros e Silva
- Laboratório de Desenvolvimento e Síntese de Fármacos, Departamento de Farmácia, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil;
| | - Natália Ferreira de Sousa
- Programa de Pós Graduação em Produtos Naturais, Sintéticos e Bioativos, Universidade Federal da Paraiba, João Pessoa 58051-900, PB, Brazil;
| | - Cristiane Flora Villarreal
- Instituto Gonçalo Moniz, Fundação Osvaldo Cruz, Salvador 40296-710, BA, Brazil; (R.F.d.E.S.); (C.F.V.)
- Faculdade de Farmácia, Universidade Federal da Bahia, Salvador 40170-290, BA, Brazil
| | - Ricardo Olímpio de Moura
- Programa de Pós Graduação em Ciências Farmacêuticas, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil; (P.R.d.S.); camillamello-@hotmail.com (C.d.O.M.); (R.O.d.M.)
- Laboratório de Desenvolvimento e Síntese de Fármacos, Departamento de Farmácia, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil;
| | - Vanda Lucia dos Santos
- Programa de Pós Graduação em Ciências Farmacêuticas, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil; (P.R.d.S.); camillamello-@hotmail.com (C.d.O.M.); (R.O.d.M.)
- Laboratório de Ensaios Farmacológicos, Departamento de Farmácia, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil; (F.E.P.C.); (T.B.C.); (Y.V.B.)
| |
Collapse
|
6
|
Greiner T, Kipp M. What Guides Peripheral Immune Cells into the Central Nervous System? Cells 2021; 10:cells10082041. [PMID: 34440810 PMCID: PMC8392645 DOI: 10.3390/cells10082041] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/05/2021] [Accepted: 08/06/2021] [Indexed: 11/16/2022] Open
Abstract
Multiple sclerosis (MS), an immune-mediated demyelinating disease of the central nervous system (CNS), initially presents with a relapsing-remitting disease course. During this early stage of the disease, leukocytes cross the blood–brain barrier to drive the formation of focal demyelinating plaques. Disease-modifying agents that modulate or suppress the peripheral immune system provide a therapeutic benefit during relapsing-remitting MS (RRMS). The majority of individuals with RRMS ultimately enter a secondary progressive disease stage with a progressive accumulation of neurologic deficits. The cellular and molecular basis for this transition is unclear and the role of inflammation during the secondary progressive disease stage is a subject of intense and controversial debate. In this review article, we discuss the following main hypothesis: during both disease stages, peripheral immune cells are triggered by CNS-intrinsic stimuli to invade the brain parenchyma. Furthermore, we outline the different neuroanatomical routes by which peripheral immune cells might migrate from the periphery into the CNS.
Collapse
|
7
|
ElSaadani M, Ahmed SM, Jacovides C, Lopez A, Johnson VE, Kaplan LJ, Smith DH, Pascual JL. Post-traumatic brain injury antithrombin III recovers Morris water maze cognitive performance, improving cued and spatial learning. J Trauma Acute Care Surg 2021; 91:108-113. [PMID: 33605694 PMCID: PMC8528176 DOI: 10.1097/ta.0000000000003112] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Neuroinflammation and cerebral edema development following severe traumatic brain injury (TBI) affect subsequent cognitive recovery. Independent of its anticoagulant effects, antithrombin III (AT-III) has been shown to block neurovascular inflammation after severe TBI, reduce cerebral endothelial-leukocyte interactions, and decrease blood-brain barrier permeability. We hypothesized that AT-III administration after TBI would improve post-TBI cognitive recovery, specifically enhancing learning, and memory. METHODS Fifteen CD1 male mice were randomized to undergo severe TBI (controlled cortical impact [CCI]: velocity, 6 m/s; depth, 1 mm; diameter, 3 mm) or sham craniotomy and received either intravenous AT-III (250 IU/kg) or vehicle (VEH/saline) 15 minutes and 24 hours post-TBI. Animals underwent Morris water maze testing from 6 to 14 days postinjury consisting of cued learning trials (platform visible), spatial learning trials (platform invisible, spatial cues present), and probe (memory) trials (platform removed, spatial cues present). Intergroup differences were assessed by the Kruskal-Wallis test (p < 0.05). RESULTS Morris water maze testing demonstrated that cumulative cued learning (overall mean time in seconds to reach the platform on days 6-8) was worst in CCI-VEH animals (26.1 ± 2.4 seconds) compared with CCI-AT-III counterparts (20.3 ± 2.1 seconds, p < 0.01). Cumulative noncued spatial learning was also worst in the CCI-VEH group (23.4 ± 1.8 seconds) but improved with AT-III (17.6 ± 1.5 seconds, p < 0.01). In probe trials, AT-III failed to significantly improve memory ability. Animals that underwent sham craniotomy demonstrated preserved learning and memory compared with all CCI counterparts (p < 0.05). CONCLUSION Antithrombin III improves neurocognitive recovery weeks after TBI. This improvement is particularly related to improvement in learning but not memory function. Pharmacologic support of enhanced learning may support new skill acquisition or relearning to improve outcomes after TBI. LEVEL OF EVIDENCE Therapeutic/care management, level II.
Collapse
Affiliation(s)
- Mohamed ElSaadani
- Division of Traumatology, Surgical Critical Care and Emergency Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Syed M. Ahmed
- Division of Traumatology, Surgical Critical Care and Emergency Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Christina Jacovides
- Division of Traumatology, Surgical Critical Care and Emergency Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Alfonso Lopez
- Division of Traumatology, Surgical Critical Care and Emergency Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Victoria E. Johnson
- Department of Neurosurgery, Center for Brain Injury, and Repair at the University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Lewis J. Kaplan
- Division of Traumatology, Surgical Critical Care and Emergency Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Douglas H. Smith
- Department of Neurosurgery, Center for Brain Injury, and Repair at the University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Jose L. Pascual
- Division of Traumatology, Surgical Critical Care and Emergency Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
- Department of Neurosurgery, Center for Brain Injury, and Repair at the University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
8
|
Akkari L, Bowman RL, Tessier J, Klemm F, Handgraaf SM, de Groot M, Quail DF, Tillard L, Gadiot J, Huse JT, Brandsma D, Westerga J, Watts C, Joyce JA. Dynamic changes in glioma macrophage populations after radiotherapy reveal CSF-1R inhibition as a strategy to overcome resistance. Sci Transl Med 2021; 12:12/552/eaaw7843. [PMID: 32669424 DOI: 10.1126/scitranslmed.aaw7843] [Citation(s) in RCA: 208] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 01/31/2020] [Accepted: 05/26/2020] [Indexed: 12/14/2022]
Abstract
Tumor-associated macrophages (TAMs) and microglia (MG) are potent regulators of glioma development and progression. However, the dynamic alterations of distinct TAM populations during the course of therapeutic intervention, response, and recurrence have not yet been fully explored. Here, we investigated how radiotherapy changes the relative abundance and phenotypes of brain-resident MG and peripherally recruited monocyte-derived macrophages (MDMs) in glioblastoma. We identified radiation-specific, stage-dependent MG and MDM gene expression signatures in murine gliomas and confirmed altered expression of several genes and proteins in recurrent human glioblastoma. We found that targeting these TAM populations using a colony-stimulating factor-1 receptor (CSF-1R) inhibitor combined with radiotherapy substantially enhanced survival in preclinical models. Our findings reveal the dynamics and plasticity of distinct macrophage populations in the irradiated tumor microenvironment, which has translational relevance for enhancing the efficacy of standard-of-care treatment in gliomas.
Collapse
Affiliation(s)
- Leila Akkari
- Department of Oncology, University of Lausanne, 1011 Lausanne, Switzerland. .,Ludwig Institute for Cancer Research, 1011 Lausanne, Switzerland.,Tumor Biology and Immunology Division, Netherlands Cancer Institute, Oncode Institute, Amsterdam 1066CX, Netherlands
| | - Robert L Bowman
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jeremy Tessier
- Tumor Biology and Immunology Division, Netherlands Cancer Institute, Oncode Institute, Amsterdam 1066CX, Netherlands
| | - Florian Klemm
- Department of Oncology, University of Lausanne, 1011 Lausanne, Switzerland.,Ludwig Institute for Cancer Research, 1011 Lausanne, Switzerland
| | - Shanna M Handgraaf
- Tumor Biology and Immunology Division, Netherlands Cancer Institute, Oncode Institute, Amsterdam 1066CX, Netherlands
| | - Marnix de Groot
- Tumor Biology and Immunology Division, Netherlands Cancer Institute, Oncode Institute, Amsterdam 1066CX, Netherlands
| | - Daniela F Quail
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Lucie Tillard
- Department of Oncology, University of Lausanne, 1011 Lausanne, Switzerland.,Ludwig Institute for Cancer Research, 1011 Lausanne, Switzerland
| | - Jules Gadiot
- Tumor Biology and Immunology Division, Netherlands Cancer Institute, Oncode Institute, Amsterdam 1066CX, Netherlands
| | - Jason T Huse
- Departments of Pathology and Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Dieta Brandsma
- Departments of Neuro-Oncology and Pathology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam 1066CX, Netherlands
| | - Johan Westerga
- Departments of Neuro-Oncology and Pathology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam 1066CX, Netherlands
| | - Colin Watts
- Birmingham Brain Cancer Program, Institute of Cancer Genome Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Johanna A Joyce
- Department of Oncology, University of Lausanne, 1011 Lausanne, Switzerland. .,Ludwig Institute for Cancer Research, 1011 Lausanne, Switzerland
| |
Collapse
|
9
|
The effects of Taurine supplementation on inflammatory markers and clinical outcomes in patients with traumatic brain injury: a double-blind randomized controlled trial. Nutr J 2021; 20:53. [PMID: 34103066 PMCID: PMC8186362 DOI: 10.1186/s12937-021-00712-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 05/25/2021] [Indexed: 11/18/2022] Open
Abstract
Background Traumatic brain injury is a public health concern and is the main cause of death among various types of trauma. The inflammatory conditions due to TBI are associated with unfavorable clinical outcomes. Taurine has been reported to have immune-modulatory effects. Thus, the aim of this study was to survey the effect of taurine supplementation in TBI patients. Methods In this study, 32 patients with TBI were randomized into two groups. The treatment group received 30 mg/kg/day of taurine in addition to the Standard Entera Meal and the control group received Standard Entera Meal for 14 days. Prior to and following the intervention, the patients were investigated in terms of serum levels of IL-6, IL-10, hs-CRP and TNF-α as well as APACHEII, SOFA and NUTRIC scores, Glasgow coma scale and weight. In addition, the length of Intensive Care Unit stay, days of dependence on ventilator and 30-day mortality were studied. SPSS software (version 13.0) was used for data analysis. Results Taurine significantly decreased the serum levels of IL-6 (p = 0.04) and marginally APACHEII score (p = 0.05). In addition, weight loss was significantly lower in taurine group (p = 0.03). Furthermore, taurine significantly increased the GCS (p = 0.03). The groups were not different significantly in terms of levels of IL-10, hs-CRP, and TNF-α, SOFA and NUTRIC scores, 30-day mortality, length of ICU stay and days of dependence on ventilator. Conclusion According to the results of the present study, taurine supplementation can reduce the IL-6 levels as one of the important inflammatory markers in these patients; and enhances the clinical outcomes too. Trial registration IRCT, IRCT20180514039657N1. Registered 22 June 2018.
Collapse
|
10
|
Revisiting Traumatic Brain Injury: From Molecular Mechanisms to Therapeutic Interventions. Biomedicines 2020; 8:biomedicines8100389. [PMID: 33003373 PMCID: PMC7601301 DOI: 10.3390/biomedicines8100389] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 09/25/2020] [Accepted: 09/26/2020] [Indexed: 12/15/2022] Open
Abstract
Studying the complex molecular mechanisms involved in traumatic brain injury (TBI) is crucial for developing new therapies for TBI. Current treatments for TBI are primarily focused on patient stabilization and symptom mitigation. However, the field lacks defined therapies to prevent cell death, oxidative stress, and inflammatory cascades which lead to chronic pathology. Little can be done to treat the mechanical damage that occurs during the primary insult of a TBI; however, secondary injury mechanisms, such as inflammation, blood-brain barrier (BBB) breakdown, edema formation, excitotoxicity, oxidative stress, and cell death, can be targeted by therapeutic interventions. Elucidating the many mechanisms underlying secondary injury and studying targets of neuroprotective therapeutic agents is critical for developing new treatments. Therefore, we present a review on the molecular events following TBI from inflammation to programmed cell death and discuss current research and the latest therapeutic strategies to help understand TBI-mediated secondary injury.
Collapse
|
11
|
Dehghanian F, Soltani Z, Khaksari M. Can Mesenchymal Stem Cells Act Multipotential in Traumatic Brain Injury? J Mol Neurosci 2020; 70:677-688. [PMID: 31897971 DOI: 10.1007/s12031-019-01475-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/26/2019] [Indexed: 12/22/2022]
Abstract
Traumatic brain injury (TBI), a leading cause of morbidity and mortality throughout the world, will probably become the third cause of death in the world by the year 2020. Lack of effective treatments approved for TBI is a major health problem. TBI is a heterogeneous disease due to the different mechanisms of injury. Therefore, it requires combination therapies or multipotential therapy that can affect multiple targets. In recent years, mesenchymal stem cells (MSCs) transplantation has considered one of the most promising therapeutic strategies to repair of brain injuries including TBI. In these studies, it has been shown that MSCs can migrate to the site of injury and differentiate into the cells secreting growth factors and anti-inflammatory cytokines. The reduction in brain edema, neuroinflammation, microglia accumulation, apoptosis, ischemia, the improvement of motor and cognitive function, and the enhancement in neurogenesis, angiogenesis, and neural stem cells survival, proliferation, and differentiation have been indicated in these studies. However, translation of MSCs research in TBI into a clinical setting will require additional preclinical trials.
Collapse
Affiliation(s)
- Fatemeh Dehghanian
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
- Bam University of Medical Sciences, Bam, Iran
| | - Zahra Soltani
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| | - Mohammad Khaksari
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
12
|
Conklin DJ, Schick S, Blaha MJ, Carll A, DeFilippis A, Ganz P, Hall ME, Hamburg N, O'Toole T, Reynolds L, Srivastava S, Bhatnagar A. Cardiovascular injury induced by tobacco products: assessment of risk factors and biomarkers of harm. A Tobacco Centers of Regulatory Science compilation. Am J Physiol Heart Circ Physiol 2019; 316:H801-H827. [PMID: 30707616 PMCID: PMC6483019 DOI: 10.1152/ajpheart.00591.2018] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 01/09/2019] [Accepted: 01/27/2019] [Indexed: 02/07/2023]
Abstract
Although substantial evidence shows that smoking is positively and robustly associated with cardiovascular disease (CVD), the CVD risk associated with the use of new and emerging tobacco products, such as electronic cigarettes, hookah, and heat-not-burn products, remains unclear. This uncertainty stems from lack of knowledge on how the use of these products affects cardiovascular health. Cardiovascular injury associated with the use of new tobacco products could be evaluated by measuring changes in biomarkers of cardiovascular harm that are sensitive to the use of combustible cigarettes. Such cardiovascular injury could be indexed at several levels. Preclinical changes contributing to the pathogenesis of disease could be monitored by measuring changes in systemic inflammation and oxidative stress, organ-specific dysfunctions could be gauged by measuring endothelial function (flow-mediated dilation), platelet aggregation, and arterial stiffness, and organ-specific injury could be evaluated by measuring endothelial microparticles and platelet-leukocyte aggregates. Classical risk factors, such as blood pressure, circulating lipoproteins, and insulin resistance, provide robust estimates of risk, and subclinical disease progression could be followed by measuring coronary artery Ca2+ and carotid intima-media thickness. Given that several of these biomarkers are well-established predictors of major cardiovascular events, the association of these biomarkers with the use of new and emerging tobacco products could be indicative of both individual and population-level CVD risk associated with the use of these products. Differential effects of tobacco products (conventional vs. new and emerging products) on different indexes of cardiovascular injury could also provide insights into mechanisms by which they induce cardiovascular harm.
Collapse
Affiliation(s)
- Daniel J Conklin
- Diabetes and Obesity Center, University of Louisville , Louisville, Kentucky
| | - Suzaynn Schick
- Department of Medicine, University of California-San Francisco , San Francisco, California
| | - Michael J Blaha
- Ciccarone Center for the Prevention of Heart Disease, Department of Medicine, Johns Hopkins University , Baltimore, Maryland
| | - Alex Carll
- Diabetes and Obesity Center, University of Louisville , Louisville, Kentucky
| | - Andrew DeFilippis
- Diabetes and Obesity Center, University of Louisville , Louisville, Kentucky
| | - Peter Ganz
- Department of Medicine, University of California-San Francisco , San Francisco, California
| | - Michael E Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
| | - Naomi Hamburg
- Department of Medicine/Cardiovascular Medicine, School of Medicine, Boston University , Boston, Massachusetts
| | - Tim O'Toole
- Diabetes and Obesity Center, University of Louisville , Louisville, Kentucky
| | - Lindsay Reynolds
- Department of Epidemiology and Prevention, Wake Forest School of Medicine , Winston-Salem, North Carolina
| | - Sanjay Srivastava
- Diabetes and Obesity Center, University of Louisville , Louisville, Kentucky
| | - Aruni Bhatnagar
- Diabetes and Obesity Center, University of Louisville , Louisville, Kentucky
| |
Collapse
|
13
|
Suto Y, Nagata K, Ahmed SM, Jacovides CL, Browne KD, Cognetti J, Johnson VE, Leone R, Kaplan LJ, Smith DH, Pascual JL. Cerebral Edema and Neurological Recovery after Traumatic Brain Injury Are Worsened if Accompanied by a Concomitant Long Bone Fracture. J Neurotrauma 2018; 36:609-618. [PMID: 30084745 DOI: 10.1089/neu.2018.5812] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Progression of severe traumatic brain injury (TBI) is associated with worsening cerebral inflammation, but it is unknown how a concomitant bone fracture (FX) affects this progression. Enoxaparin (ENX), a low molecular weight heparin often used for venous thromboembolic prophylaxis, decreases penumbral leukocyte (LEU) mobilization in isolated TBI and improves neurological recovery. We investigated if TBI accompanied by an FX worsens LEU-mediated cerebral inflammation and if ENX alters this process. CD1 male mice underwent controlled cortical impact (CCI) or sham craniotomy with or without an open tibial FX, and received either ENX (1 mg/kg, three times/day) or saline for 2 days following injury. Randomization defined four groups (Sham, CCI, CCI+FX, CCI+FX+ENX, n = 10/group). Two days after CCI, neurological recovery was assessed with the Garcia Neurological Test (GNT); intravital microscopy (LEU rolling and adhesion, microvascular leakage) and blood hemoglobin levels were also evaluated. Penumbral cerebral neutrophil sequestration (Ly-6G immunohistochemistry [IHC]) were evaluated post-mortem. In vivo LEU rolling was greater in CCI+FX (45.2 ± 4.8 LEUs/100 μm/min) than in CCI alone (26.5 ± 3.1, p = 0.007), and was suppressed by ENX (23.2 ± 5.5, p = 0.003 vs. CCI + FX). Neurovascular permeability was higher in CCI+FX (71.1 ± 2.9%) than CCI alone (42.5 ± 2.3, p < 0.001). GNT scores were lower in CCI+FX (15.2 ± 0.2) than in CCI alone (16.3 ± 0.3, p < 0.001). Hemoglobin was lowest in the CCI+FX+ENX group, lower than in Sham or CCI. IHC demonstrated greatest polymorphonuclear neutrophil (PMN) invasion in CCI+FX in uninjured cerebral territories. A concomitant long bone FX worsens TBI-induced cerebral LEU mobilization, microvascular leakage, and cerebral edema, and impairs neurological recovery at 48 h. ENX suppresses this progression but may increase bleeding.
Collapse
Affiliation(s)
- Yujin Suto
- 1 Division of Traumatology, Surgical Clinical Care and Emergency Surgery, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania.,2 Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania
| | - Katsuhiro Nagata
- 3 Department of Emergency and Critical Care Medicine, Tokyo Medical University Hachioji Medical Center , Tokyo, Japan
| | - Syed M Ahmed
- 1 Division of Traumatology, Surgical Clinical Care and Emergency Surgery, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania.,2 Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania
| | - Christina L Jacovides
- 1 Division of Traumatology, Surgical Clinical Care and Emergency Surgery, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania.,2 Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania
| | - Kevin D Browne
- 2 Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania
| | - John Cognetti
- 2 Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania
| | - Victoria E Johnson
- 2 Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania
| | - Ryan Leone
- 1 Division of Traumatology, Surgical Clinical Care and Emergency Surgery, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania
| | - Lewis J Kaplan
- 1 Division of Traumatology, Surgical Clinical Care and Emergency Surgery, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania
| | - Douglas H Smith
- 2 Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania
| | - Jose L Pascual
- 1 Division of Traumatology, Surgical Clinical Care and Emergency Surgery, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania.,2 Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania
| |
Collapse
|
14
|
Uz Z, van Gulik TM, Aydemirli MD, Guerci P, Ince Y, Cuppen D, Ergin B, Aksu U, de Mol BA, Ince C. Identification and quantification of human microcirculatory leukocytes using handheld video microscopes at the bedside. J Appl Physiol (1985) 2018. [PMID: 29517420 DOI: 10.1152/japplphysiol.00962.2017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Leukocyte recruitment and adhesion to the endothelium are hallmarks of systemic inflammation that manifest in a wide range of diseases. At present, no method is available to directly measure leukocyte kinetics at the bedside. In this study, we validate a new method to identify and quantify microcirculatory leukocytes observed by handheld vital microscopy (HVM) using space-time diagram (STD) analysis. Video clips ( n = 59) containing one capillary-postcapillary venule unit where leukocytes could be observed emanating from a capillary into a venule in cardiac surgery patients ( n = 20) were included. STD analysis and manual counting were used to quantify the number of leukocytes (total, rolling, and nonrolling). Pearson's correlation and Bland-Altman analysis were used to determine agreement between the STDs and manual counting. For reproducibility, intra- and interobserver coefficients of variation (CVs) were assessed. Leukocyte (rolling and nonrolling) and red blood cell velocities were assessed. The STDs and manual counting procedures for the quantification of rolling leukocytes showed good agreement ( r = 0.8197, P < 0.0001), with a Bland-Altman analysis mean difference of -0.0 (-6.56; 6.56). The overall intraobserver CV for the STD method was 1.5%. The overall interobserver CVs for the STD and the manual method were 5.6% and 9.4%, respectively. The nonrolling velocity was significantly higher than the rolling velocity (812 ± 519 µm/s vs. 201 ± 149 µm/s, P = 0.001). STD results agreed with the manual counting procedure results, had a better reproducibility, and could assess the leukocyte velocity. STD analysis using bedside HVM imaging presented a new methodology for quantifying leukocyte kinetics and functions in the microcirculation. NEW & NOTEWORTHY In this study, we introduce space-time diagram analysis of sublingual microcirculation imaging using handheld vital microscopy to identify and quantify the presence and kinetics of human microcirculatory leukocytes. We validated the methodology by choosing anatomical units consisting of a capillary connected to a venule, which allowed precise identification of leukocytes.
Collapse
Affiliation(s)
- Zühre Uz
- Department of Translational Physiology, Academic Medical Center, University of Amsterdam , Amsterdam , The Netherlands.,Department of Experimental Surgery, Academic Medical Center, University of Amsterdam , Amsterdam , The Netherlands
| | - Thomas M van Gulik
- Department of Experimental Surgery, Academic Medical Center, University of Amsterdam , Amsterdam , The Netherlands
| | - Mehtap D Aydemirli
- Department of Translational Physiology, Academic Medical Center, University of Amsterdam , Amsterdam , The Netherlands
| | - Philippe Guerci
- Department of Translational Physiology, Academic Medical Center, University of Amsterdam , Amsterdam , The Netherlands
| | - Yasin Ince
- Department of Translational Physiology, Academic Medical Center, University of Amsterdam , Amsterdam , The Netherlands
| | - Diede Cuppen
- Department of Translational Physiology, Academic Medical Center, University of Amsterdam , Amsterdam , The Netherlands
| | - Bulent Ergin
- Department of Translational Physiology, Academic Medical Center, University of Amsterdam , Amsterdam , The Netherlands.,Department of Intensive Care, Erasmus MC University Hospital Rotterdam , Rotterdam , The Netherlands
| | - Ugur Aksu
- Department of Translational Physiology, Academic Medical Center, University of Amsterdam , Amsterdam , The Netherlands.,Department of Biology, The University of Istanbul , Istanbul , Turkey
| | - Bas A de Mol
- Department of Cardio-Thoracic Surgery, Academic Medical Center, University of Amsterdam , Amsterdam , The Netherlands
| | - Can Ince
- Department of Translational Physiology, Academic Medical Center, University of Amsterdam , Amsterdam , The Netherlands.,Department of Intensive Care, Erasmus MC University Hospital Rotterdam , Rotterdam , The Netherlands
| |
Collapse
|
15
|
Cerebrovascular heterogeneity and neuronal excitability. Neurosci Lett 2018; 667:75-83. [DOI: 10.1016/j.neulet.2017.01.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 01/04/2017] [Accepted: 01/06/2017] [Indexed: 01/01/2023]
|
16
|
Nagata K, Kumasaka K, Browne KD, Li S, St-Pierre J, Cognetti J, Marks J, Johnson VE, Smith DH, Pascual JL. Unfractionated heparin after TBI reduces in vivo cerebrovascular inflammation, brain edema and accelerates cognitive recovery. J Trauma Acute Care Surg 2017; 81:1088-1094. [PMID: 27533909 DOI: 10.1097/ta.0000000000001215] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Severe traumatic brain injury (TBI) may increase the risk of venous thromboembolic complications; however, early prevention with heparinoids is often withheld for its anticoagulant effect. New evidence suggests low molecular weight heparin reduces cerebral edema and improves neurological recovery after stroke and TBI, through blunting of cerebral leukocyte (LEU) recruitment. It remains unknown if unfractionated heparin (UFH) similarly affects brain inflammation and neurological recovery post-TBI. We hypothesized that UFH after TBI reduces cerebral edema by reducing LEU-mediated inflammation and improves neurological recovery. METHODS CD1 male mice underwent either TBI by controlled cortical impact (CCI) or sham craniotomy. UFH (75 U/kg or 225 U/kg) or vehicle (VEH, 0.9% saline) was administered 2, 11, 20, 27, and 34 hours after TBI. At 48 hours, pial intravital microscopy through a craniotomy was used to visualize live brain LEUs interacting with endothelium and microvascular fluorescein isothiocyanate-albumin leakage. Neurologic function (Garcia Neurological Test, GNT) and body weight loss ratios were evaluated 24 and 48 hours after TBI. Cerebral and lung wet-to-dry ratios were evaluated post mortem. ANOVA with Bonferroni correction was used to determine significance (p < 0.05). RESULTS Compared to positive controls (CCI), both UFH doses reduced post-TBI in vivo LEU rolling on endothelium, concurrent cerebrovascular albumin leakage, and ipsilateral cerebral water content after TBI. Additionally, only low dose UFH (75 U/kg) improved GNT at both 24 and 48 hours after TBI. High dose UFH (225 U/kg) significantly increased body weight loss above sham at 48 hours. Differences in lung water content and blood pressure between groups were not significant. CONCLUSIONS UFH after TBI reduces LEU recruitment, microvascular permeability, and brain edema to injured brain. Lower UFH doses concurrently improve neurological recovery whereas higher UFH may worsen functional recovery. Further study is needed to determine if this is caused by increased bleeding from injured brain with higher UFH doses.
Collapse
Affiliation(s)
- Katsuhiro Nagata
- From the Division of Traumatology, Surgical Critical Care & Emergency Surgery (K.N., J.S-P., J.L.P.) and Department of Neurosurgery, Center for Brain Injury and Repair (K.D.B., J.C., V.E.J., D.H.S., J.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania; Sidney Kimmel Medical College at Thomas Jefferson University (J.M.), Philadelphia, Pennsylvania; Department of Neurosurgery (S.L.), Qianfoshan Hospital, Shandong University, Jinan, China; and Department of Emergency and Critical Care Medicine (K.K.), Tokyo Medical University Hachioji Medical Center, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Kunutsor SK, Bakker SJL, Dullaart RPF. Soluble Vascular Cell Adhesion Molecules May be Protective of Future Cardiovascular Disease Risk: Findings from the PREVEND Prospective Cohort Study. J Atheroscler Thromb 2017; 24:804-818. [PMID: 28202840 PMCID: PMC5556188 DOI: 10.5551/jat.38836] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Aim: Soluble cell adhesion molecules, such as vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1, E-selectin, and P-selectin, have been suggested to be associated with cardiovascular disease (CVD) risk; however, the nature and magnitude of the association between VCAM-1 and CVD risk is uncertain. We aimed to assess the association of VCAM-1 with CVD risk and determine its potential utility for CVD risk prediction. Methods: VCAM-1 concentrations were measured at baseline in the PREVEND prospective study of 2,638 participants. Hazard ratios (95% confidence intervals [CI]) and measures of risk discrimination for CVD (e.g., C-index) and reclassification (i.e., net reclassification improvement) of participants were assessed. Results: During a median follow-up of 9.9 years, 614 CVD events occurred. Plasma VCAM-1 was weakly associated with several cardiovascular risk markers. In analyses adjusted for established cardiovascular risk factors, the hazard ratio (95% CI) for CVD per 1 standard deviation increase in loge VCAM-1 was 0.91 (0.84–0.99; P = 0.020), which remained consistent after additional adjustment for body mass index, alcohol consumption, triglycerides, renal function, and C-reactive protein; hazard ratio (95% CI) 0.89 (0.82–0.97; P = 0.006). Comparing the top versus bottom quintiles of VCAM-1 levels, the corresponding adjusted hazard ratios were 0.74 (0.57–0.96; P = 0.023) and 0.70 (0.54–0.91; P = 0.007) respectively. Adding VCAM-1 to a CVD risk prediction model containing conventional risk factors did not improve the C-index or net reclassification. Conclusions: Plasma VCAM-1 is inversely and independently associated with CVD. However, VCAM-1 provides no significant improvement in CVD risk assessment beyond conventional CVD risk factors.
Collapse
Affiliation(s)
| | - Stephan J L Bakker
- Department of Nephrology Medicine, University of Groningen and University Medical Center Groningen.,Top Institute Food and Nutrition
| | - Robin P F Dullaart
- Department of Endocrinology, University of Groningen and University Medical Center Groningen
| |
Collapse
|
18
|
Pharmacological opportunities to control inflammatory diseases through inhibition of the leukocyte recruitment. Pharmacol Res 2016; 112:37-48. [DOI: 10.1016/j.phrs.2016.01.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 01/14/2016] [Accepted: 01/15/2016] [Indexed: 12/30/2022]
|
19
|
Tong H, Jiang G, Guan X, Wu H, Song K, Cheng K, Sun X. Characterization of a polysaccharide from Rosa davurica and inhibitory activity against neutrophil migration. Int J Biol Macromol 2016; 89:111-7. [PMID: 27112979 DOI: 10.1016/j.ijbiomac.2016.04.062] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 04/15/2016] [Accepted: 04/21/2016] [Indexed: 12/21/2022]
Abstract
The rapid recruitment of neutrophils from peripheral blood into infected sites is critical step for inflammatory responses; however, the excessive and improper recruitment can lead to serious tissue damages. Thus, it is a promising strategy to inhibit their excessive recruitment for treating inflammation-related disease. Here, we isolated a polysaccharide (RDPA1) from Rosa davurica, to evaluate its physicochemical property and inhibitory effects on neutrophil migration. RDPA1 was obtained by hot-water extraction, ethanol precipitation, and fractionated by DEAE-cellulose and Sepharose CL-6B columns. RDPA1 significantly inhibited in vitro migration of human neutrophils evaluated by transwell chamber and impacted the migratory behavior observed by time-lapsed microscopy, we found the migrated distance and average velocity of RDPA1-treated cells were greatly reduced. In addition, RDPA1 treatment impaired in vivo neutrophil infiltration in the peritonitis mice. RDPA1 exhibited significant blocking capacity of the interaction between β2 integrins and ICAM-1 evaluated by flow cytometry and in vitro protein binding assay. Together, these results suggest RDPA1 could be considered as a potential candidate for developing a novel anti-inflammatory agent.
Collapse
Affiliation(s)
- Haibin Tong
- Jilin Provincial Key Laboratory of Molecular Geriatric Medicine, Life Science Research Center, Beihua University, Jilin 132013, China; Wood Material Science and Engineering Key Laboratory of Jilin Province, Beihua University, Jilin 132013, China
| | - Guiquan Jiang
- Wood Material Science and Engineering Key Laboratory of Jilin Province, Beihua University, Jilin 132013, China
| | - Xingang Guan
- Jilin Provincial Key Laboratory of Molecular Geriatric Medicine, Life Science Research Center, Beihua University, Jilin 132013, China
| | - Hong Wu
- Laboratory of Cell Imaging, Henan University of Traditional Chinese Medicine, Zhengzhou 450002, China
| | - Kangxing Song
- Department of Cardiology, The General Hospital of Chinese People's Liberation Army, Beijing 100853, China
| | - Kuan Cheng
- Affiliated Hospital, Northeast Normal University, Changchun 130024, China
| | - Xin Sun
- Jilin Provincial Key Laboratory of Molecular Geriatric Medicine, Life Science Research Center, Beihua University, Jilin 132013, China.
| |
Collapse
|
20
|
Enoxaparin ameliorates post-traumatic brain injury edema and neurologic recovery, reducing cerebral leukocyte endothelial interactions and vessel permeability in vivo. J Trauma Acute Care Surg 2015; 79:78-84. [PMID: 26091318 DOI: 10.1097/ta.0000000000000697] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
BACKGROUND Traumatic brain injury (TBI) confers a high risk of venous thrombosis, but early prevention with heparinoids is often withheld, fearing cerebral hematoma expansion. Yet, studies have shown heparinoids not only to be safe but also to limit brain edema and contusion size after TBI. Human TBI data also suggest faster radiologic and clinical neurologic recovery with earlier heparinoid administration. We hypothesized that enoxaparin (ENX) after TBI blunts in vivo leukocyte (LEU) mobilization to injured brain and cerebral edema, while improving neurologic recovery without increasing the size of the cerebral hemorrhagic contusion. METHODS CD1 male mice underwent either TBI by controlled cortical impact (CCI, 1-mm depth, 6 m/s) or sham craniotomy. ENX (1 mg/kg) or vehicle (VEH, 0.9% saline, 1 mL/kg) was administered at 2, 8, 14, 23, and 32 hours after TBI. At 48 hours, intravital microscopy was used to visualize live LEUs interacting with endothelium and microvascular leakage of fluorescein isothiocyanate-albumin. Neurologic function (Neurological Severity Score, NSS), activated clotting time, hemorrhagic contusion size, as well as brain and lung wet-to-dry ratios were evaluated post mortem. Analysis of variance with Bonferroni correction was used for statistical comparisons between groups. RESULTS Compared with VEH, ENX significantly reduced in vivo LEU rolling on endothelium (72.7 ± 28.3 LEU/100 μm/min vs. 30.6 ± 18.3 LEU/100 μm/min, p = 0.02) and cerebrovascular albumin leakage (34.5% ± 8.1% vs. 23.8% ± 5.5%, p = 0.047). CCI significantly increased ipsilateral cerebral hemisphere edema, but ENX treatment reduced post-CCI edema to near control levels (81.5% ± 1.5% vs. 77.6% ± 0.6%, p < 0.01). Compared with VEH, ENX reduced body weight loss at 24 hours (8.7% ± 1.2% vs. 5.8% ± 1.1%, p < 0.01) and improved NSS at 24 hours (14.5 ± 0.5 vs. 16.2 ± 0.4, p < 0.01) and 48 hours (15.1 ± 0.4 vs. 16.7 ± 0.5, p < 0.01) after injury. There were no significant differences in activated clotting time, hemorrhagic contusion size, and lung water content between the groups. CONCLUSION ENX reduces LEU recruitment to injured brain, diminishing visible microvascular permeability and edema. ENX may also accelerate neurologic recovery without increasing cerebral contusion size. Further study in humans is necessary to determine safety, appropriate dosage, and timing of ENX administration early after TBI.
Collapse
|
21
|
Tong H, Song J, Zhang Z, Mao D, Sun G, Jiang G. Inhibitory function of P-selectin-mediated leukocyte adhesion by the polysaccharides from Sanguisorba officinalis. PHARMACEUTICAL BIOLOGY 2015; 53:345-349. [PMID: 25331826 DOI: 10.3109/13880209.2014.919597] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
CONTEXT P-selectin is a promising target for inflammatory-related diseases. Polysaccharides are the active ingredients of Sanguisorba officinalis L. (Rosaceae) responsible for its anti-inflammatory activities; however, the molecular mechanism is not clear yet. OBJECTIVE This study evaluates the effects of polysaccharides (SOPs) from Sanguisorba officinalis on their antagonistic function against P-selectin-mediated leukocyte adhesion. MATERIALS AND METHODS The antagonistic function of SOPs was investigated by flow cytometry and static adhesion assay at the concentrations of 25 and 100 μg/ml. The dynamic interaction between HL-60 cells and CHO-P cell monolayer treated with SOPs (25 and 100 μg/ml) was analyzed in a parallel plate flow chamber, and quantitatively calculated by ImageJ software (NIH, Bethesda, MD). In vitro protein binding assay was carried out to evaluate the blocking effects of SOPs (25 and 100 μg/ml) on the interaction between P-selectin and PSGL-1. RESULTS SOPs-treatment (100 μg/ml) significantly reduced the percentage of HL-60 cells binding to P-selectin (p < 0.01) determined by flow cytometry. In addition, SOPs (25 and 100 μg/ml) markedly blocked the adhesion between HL-60 cells and CHO-P cells under static condition, and the inhibitory rates reached 39.9% and 71.2%, respectively. Compared with the positive control group, SOPs-treatment (25 and 100 μg/ml) significantly reduced the percentage of HL-60 cells rolling on CHO-P cell monolayers by 43.5% and 75.2%, respectively. Protein binding assay showed the interaction between P-selectin and PSGL-1 was significantly blocked by SOPs. DISCUSSION AND CONCLUSION SOPs possess a significant antagonistic function against P-selectin-mediated leukocyte adhesion, and SOPs could be considered as a promising candidate for amelioration of inflammation-related diseases.
Collapse
Affiliation(s)
- Haibin Tong
- College of Forestry, Beihua University , Jilin , China
| | | | | | | | | | | |
Collapse
|
22
|
Inhibition of inflammatory injure by polysaccharides from Bupleurum chinense through antagonizing P-selectin. Carbohydr Polym 2014; 105:20-5. [PMID: 24708947 DOI: 10.1016/j.carbpol.2014.01.039] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 01/10/2014] [Accepted: 01/12/2014] [Indexed: 12/21/2022]
Abstract
P-selectin-mediated adhesion between endothelium and neutrophils is a crucial process leading to acute inflammatory injure. Thus, P-selectin has been considered as promising target for therapeutics of acute inflammatory-related diseases. In the present study, the water-soluble polysaccharides (BCPs) were isolated from Bupleurum chinense, and we evaluated their therapeutical effects on acute inflammatory injure and antagonistic function against P-selectin-mediated neutrophil adhesion. Our results showed that BCPs significantly impaired the leukocyte infiltration and relieve lung injury in LPS-induced acute pneumonia model. BCPs significantly blocked the binding of P-selectin to neutrophils and inhibited P-selectin-mediated neutrophils rolling along CHO-P cell monolayer. The result from in vitro protein binding assay showed a direct evidence indicating that BCPs-treatment significantly eliminated the interaction between rhP-Fc and its physiological ligand PSGL-1 at protein level. Together, these results provide a novel therapeutical strategy for amelioration of inflammation-related disease processes by polysaccharides from B. chinense.
Collapse
|
23
|
Traffic of Leukocytes and Cytokine Up-regulation in the Central Nervous System in a Murine Model of Neuroparacoccidioidomycosis. Mycopathologia 2013; 176:191-9. [DOI: 10.1007/s11046-013-9679-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 07/08/2013] [Indexed: 02/06/2023]
|
24
|
Mayer CL, Huber BR, Peskind E. Traumatic brain injury, neuroinflammation, and post-traumatic headaches. Headache 2013; 53:1523-30. [PMID: 24090534 DOI: 10.1111/head.12173] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2013] [Indexed: 12/12/2022]
Abstract
Concussions following head and/or neck injury are common, and although most people with mild injuries recover uneventfully, a subset of individuals develop persistent post-concussive symptoms that often include headaches. Post-traumatic headaches vary in presentation and may progress to become chronic and in some cases debilitating. Little is known about the pathogenesis of post-traumatic headaches, although shared pathophysiology with that of the brain injury is suspected. Following primary injury to brain tissues, inflammation rapidly ensues; while this inflammatory response initially provides a defensive/reparative function, it can persist beyond its beneficial effect, potentially leading to secondary injuries because of alterations in neuronal excitability, axonal integrity, central processing, and other changes. These changes may account for the neurological symptoms often observed after traumatic brain injury, including headaches. This review considers selected aspects of the inflammatory response following traumatic brain injury, with an emphasis on the role of glial cells as mediators of maladaptive post-traumatic inflammation.
Collapse
Affiliation(s)
- Cynthia L Mayer
- VA Northwest Network Mental Illness Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, USA; Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA
| | | | | |
Collapse
|
25
|
Inflammatory biomarkers for predicting cardiovascular disease. Clin Biochem 2013; 46:1353-71. [PMID: 23756129 DOI: 10.1016/j.clinbiochem.2013.05.070] [Citation(s) in RCA: 132] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Revised: 05/27/2013] [Accepted: 05/30/2013] [Indexed: 02/07/2023]
Abstract
The pathology of cardiovascular disease (CVD) is complex; multiple biological pathways have been implicated, including, but not limited to, inflammation and oxidative stress. Biomarkers of inflammation and oxidative stress may serve to help identify patients at risk for CVD, to monitor the efficacy of treatments, and to develop new pharmacological tools. However, due to the complexities of CVD pathogenesis there is no single biomarker available to estimate absolute risk of future cardiovascular events. Furthermore, not all biomarkers are equal; the functions of many biomarkers overlap, some offer better prognostic information than others, and some are better suited to identify/predict the pathogenesis of particular cardiovascular events. The identification of the most appropriate set of biomarkers can provide a detailed picture of the specific nature of the cardiovascular event. The following review provides an overview of existing and emerging inflammatory biomarkers, pro-inflammatory cytokines, anti-inflammatory cytokines, chemokines, oxidative stress biomarkers, and antioxidant biomarkers. The functions of each biomarker are discussed, and prognostic data are provided where available.
Collapse
|
26
|
Defibrotide: properties and clinical use of an old/new drug. Vascul Pharmacol 2013; 59:1-10. [PMID: 23680861 DOI: 10.1016/j.vph.2013.05.001] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 04/29/2013] [Accepted: 05/03/2013] [Indexed: 12/30/2022]
Abstract
The drug named defibrotide (DFT) has been studied for many years. It has been shown to possess many activities: profibrinolytic, antithrombotic-thrombolytic, antiischemic (heart, liver, kidney, skin, brain), antishock, antiatherosclerotic, antirejection and anti-angiogenic. The previously displayed activities, as antithrombotic, profibrinolytic and anti-inflammatory, suggested its use in vascular disorders, as in the treatment of peripheral obliterative arterial disease and in thrombophlebitis. Some years after, the use of DFT in hepatic veno-occlusive disease has been also proposed. Even if DFT was considered for long time a multi-target drug, now it could be considered on the whole as a drug able to protect endothelium against activation. The present work reviews the more important experimental and clinical studies performed to detect DFT effects.
Collapse
|
27
|
Vilela MC, Lima GK, Rodrigues DH, Lacerda-Queiroz N, Pedroso VSP, Miranda AS, Rachid MA, Kroon EG, Campos MA, Teixeira MM, Sellner J, Teixeira AL. Absence of CCR5 increases neutrophil recruitment in severe herpetic encephalitis. BMC Neurosci 2013; 14:19. [PMID: 23391218 PMCID: PMC3618319 DOI: 10.1186/1471-2202-14-19] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 01/30/2013] [Indexed: 12/31/2022] Open
Abstract
Background The neuroinflammatory response aimed at clearance of herpes simplex virus-1 (HSV-1) plays a key role in the pathogenesis of neuroaxonal damage in herpetic encephalitis. Leukocytes activated in an adaptive immune response access brain tissue by passing through the blood–brain barrier. The chemokine CCL5/RANTES is involved in recruitment of these cells to the brain acting via the receptors CCR1, CCR3 and mainly CCR5. Here, we evaluated the role of CCR5 on traffic of leukocytes in the brain microvasculature, cellular and cytokines profile in a severe form of herpetic encephalitis. Results Wild type and mice lacking CCR5 (CCR5-/-) were inoculated intracerebrally with 104 PFU of neurotropic HSV-1. We evaluated the traffic of leukocytes in the brain microvasculature using intravital microscopy and the profile of cytokines by Enzyme-Linked Immunosorbent Assay at 1 day post infection. Flow cytometry and histopathological analyses were also carried out in brain tissue. Absence of CCR5 leads to lower viral load and an increased leukocyte adhesion in brain microvasculature, predominantly of neutrophils (CD11+ Ly6G+ cells). Moreover, there was a significant increase in the levels of MIP-1/CCL2, RANTES/CCL5, KC/CXCL1 and MIG/CXCL9 in the brain of infected CCR5-/- mice. Conclusions These results suggest that the absence of CCR5 may boost the immune response with a high neutrophil recruitment which most likely helps in viral clearance. Nonetheless, the elevated immune response may be detrimental to the host.
Collapse
Affiliation(s)
- Márcia Carvalho Vilela
- Pós-Graduação em Ciências da Saúde, Infectologia e Medicina Tropical, Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antonio Carlos, 6627 Pampulha, Belo Horizonte, MG 31270-901, Brazil.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Attenuating inflammation but stimulating both angiogenesis and neurogenesis using hyperbaric oxygen in rats with traumatic brain injury. J Trauma Acute Care Surg 2012; 72:650-9. [PMID: 22491549 DOI: 10.1097/ta.0b013e31823c575f] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Inflammation, angiogenesis, neurogenesis, and gliosis are involved in traumatic brain injury (TBI). Several studies provide evidence supporting the neuroprotective effect of hyperbaric oxygen (HBO2) therapy in TBI. The aim of this study was to ascertain whether inflammation, angiogenesis, neurogenesis, and gliosis during TBI are affected by HBO2 therapy. METHODS Rats were randomly divided into three groups: TBI + NBA (normobaric air: 21% O2 at 1 absolute atmospheres), TBI + HBO2, and Sham operation + NBA. TBI + HBO2 rats received 100% O2 at 2.0 absolute atmospheres for 1 hr/d for three consecutive days. Behavioral tests and biochemical and histologic evaluations were done 4 days after TBI onset. RESULTS TBI + NBA rats displayed: (1) motor and cognitive dysfunction; (2) cerebral infarction and apoptosis; (3) activated inflammation (evidenced by increased brain myeloperoxidase activity and higher serum levels of tumor necrosis factor-α); (4) neuronal loss (evidenced by fewer NeuN-positive cells); and (5) gliosis (evidenced by more glial fibrillary protein-positive cells). In TBI + HBO2 rats, HBO2 therapy significantly reduced TBI-induced motor and cognitive dysfunction, cerebral infarction and apoptosis, activated inflammation, neuronal loss, and gliosis. In addition, HBO2 therapy stimulated angiogenesis (evidenced by more bromodeoxyuridine-positive endothelial and vascular endothelial growth factor-positive cells), neurogenesis (evidenced by more bromodeoxyuridine-NeuN double-positive and glial cells-derived neurotrophic factor-positive cells), and overproduction of interleukin-10 (an anti-inflammatory cytokine). CONCLUSIONS Collectively, these results suggest that HBO2 therapy may improve outcomes of TBI in rats by inhibiting activated inflammation and gliosis while stimulating both angiogenesis and neurogenesis in the early stage.
Collapse
|
29
|
Vilela MC, Campos RDDL, Mansur DS, Rodrigues DH, Lacerda-Queiroz N, Lima GK, Rachid MA, Kroon EG, Campos MA, Teixeira AL. Role of IL-4 in an experimental model of encephalitis induced by intracranial inoculation of herpes simplex virus-1 (HSV-1). ARQUIVOS DE NEURO-PSIQUIATRIA 2011; 69:237-41. [PMID: 21537568 DOI: 10.1590/s0004-282x2011000200019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Accepted: 10/20/2010] [Indexed: 01/30/2023]
Abstract
Herpes simplex virus-1 (HSV-1) is a pathogen that may cause severe encephalitis in humans. In this study, we aimed to investigate the role of interleukin-4 (IL-4) in a model of HSV-1 brain infection. IL-4 knockout (IL-4-/-) and wild type (WT) C57BL/6 mice were inoculated with 10(4) plaque-forming units of HSV-1 by the intracranial route. Histopathologic analysis revealed a distinct profile of infiltrating cells at 3 days post-infection (dpi). Infected WT mice presented mononuclear inflammatory cells while IL-4-/- mice developed meningoencephalitis with predominance of neutrophils. IL-4-/- mice had diminished leukocyte adhesion at 3 dpi when compared to infected WT animals in intravital microscopy study. Conversely no differences were found in cerebral levels of CXCL1, CXCL9, CCL3, CCL5 and TNF-α between WT and IL-4-/- infected mice. IL-4 may play a role in the recruitment of cells into central nervous system in this acute model of severe encephalitis caused by HSV-1.
Collapse
Affiliation(s)
- Márcia Carvalho Vilela
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, UFMG, Belo Horizonte, MG, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Ifergan I, Kebir H, Alvarez JI, Marceau G, Bernard M, Bourbonnière L, Poirier J, Duquette P, Talbot PJ, Arbour N, Prat A. Central nervous system recruitment of effector memory CD8+ T lymphocytes during neuroinflammation is dependent on α4 integrin. Brain 2011; 134:3560-77. [PMID: 22058139 PMCID: PMC7110084 DOI: 10.1093/brain/awr268] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Clonally expanded CD8+ T lymphocytes are present in multiple sclerosis lesions, as well as in the cerebrospinal fluid of patients with multiple sclerosis. In experimental autoimmune encephalomyelitis, CD8+ T lymphocytes are found in spinal cord and brainstem lesions. However, the exact phenotype of central nervous system-infiltrating CD8+ T lymphocytes and the mechanism by which these cells cross the blood–brain barrier remain largely unknown. Using cerebrospinal fluid from patients with multiple sclerosis, spinal cord from experimental autoimmune encephalomyelitis and coronavirus-induced encephalitis, we demonstrate that central nervous system-infiltrating CD8+ T lymphocytes are mostly of the effector memory phenotype (CD62L− CCR7− granzymeBhi). We further show that purified human effector memory CD8+ T lymphocytes transmigrate more readily across blood-brain barrier-endothelial cells than non-effector memory CD8+ T lymphocytes, and that blood-brain barrier endothelium promotes the selective recruitment of effector memory CD8+ T lymphocytes. Furthermore, we provide evidence for the recruitment of interferon-γ- and interleukin-17-secreting CD8+ T lymphocytes by human and mouse blood-brain barrier endothelium. Finally, we show that in vitro migration of CD8+ T lymphocytes across blood-brain barrier-endothelial cells is dependent on α4 integrin, but independent of intercellular adhesion molecule-1/leucocyte function-associated antigen-1, activated leucocyte cell adhesion molecule/CD6 and the chemokine monocyte chemotactic protein-1/CCL2. We also demonstrate that in vivo neutralization of very late antigen-4 restricts central nervous system infiltration of CD8+ T lymphocytes in active immunization and adoptive transfer experimental autoimmune encephalomyelitis, and in coronavirus-induced encephalitis. Our study thus demonstrates an active role of the blood-brain barrier in the recruitment of effector memory CD8+ T lymphocytes to the CNS compartment and defines α4 integrin as a major contributor of CD8+ T lymphocyte entry into the brain.
Collapse
Affiliation(s)
- Igal Ifergan
- Neuroimmunology Research Unit, Centre for Excellence in Neuromics, CRCHUM-Notre-Dame Hospital, Université de Montréal, Montréal, QC, H2L 4M1, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Structural characterization and in vitro inhibitory activities in P-selectin-mediated leukocyte adhesion of polysaccharide fractions isolated from the roots of Physalis alkekengi. Int J Biol Macromol 2011; 49:210-7. [DOI: 10.1016/j.ijbiomac.2011.04.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Revised: 04/11/2011] [Accepted: 04/22/2011] [Indexed: 11/16/2022]
|
32
|
Guerrieri D, Tateosian NL, Maffía PC, Reiteri RM, Amiano NO, Costa MJ, Villalonga X, Sanchez ML, Estein SM, Garcia VE, Sallenave JM, Chuluyan HE. Serine leucocyte proteinase inhibitor-treated monocyte inhibits human CD4(+) lymphocyte proliferation. Immunology 2011; 133:434-41. [PMID: 21574992 DOI: 10.1111/j.1365-2567.2011.03451.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Serine leucocyte proteinase inhibitor (SLPI) is the main serine proteinase inhibitor produced by epithelial cells and has been shown to be a pleiotropic molecule with anti-inflammatory and microbicidal activities. However, the role of SLPI on the adaptive immune response is not well established. Therefore, we evaluated the effect of SLPI on lymphocyte proliferation and cytokine production. Human peripheral blood mononuclear cells (PBMC) were treated with mitogens plus SLPI and proliferation was assessed by [(3) H]thymidine uptake. The SLPI decreased the lymphocyte proliferation induced by interleukin-2 (IL-2) or OKT3 monoclonal antibodies in a dose-dependent manner. Inhibition was not observed when depleting monocytes from the PBMC and it was restored by adding monocytes and SLPI. SLPI-treated monocyte slightly decreased MHC II and increased CD18 expression, and secreted greater amounts of IL-4, IL-6 and IL-10 in the cell culture supernatants. SLPI-treated monocyte culture supernatant inhibited the CD4(+) lymphocyte proliferation but did not affect the proliferation of CD8(+) cells. Moreover, IL-2 increased T-bet expression and the presence of SLPI significantly decreased it. Finally, SLPI-treated monocyte culture supernatant dramatically decreased interferon-γ but increased IL-4, IL-6 and IL-10 in the presence of IL-2-treated T cells. Our results demonstrate that SLPI target monocytes, which in turn inhibit CD4 lymphocyte proliferation and T helper type 1 cytokine secretion. Overall, these results suggest that SLPI is an alarm protein that modulates not only the innate immune response but also the adaptive immune response.
Collapse
Affiliation(s)
- Diego Guerrieri
- Departamento de Farmacología, Universidad de Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
von Wedel-Parlow M, Schrot S, Lemmen J, Treeratanapiboon L, Wegener J, Galla HJ. Neutrophils cross the BBB primarily on transcellular pathways: an in vitro study. Brain Res 2010; 1367:62-76. [PMID: 20875807 DOI: 10.1016/j.brainres.2010.09.076] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Accepted: 09/21/2010] [Indexed: 01/13/2023]
Abstract
The cerebral microcapillary endothelium forms a highly important barrier between the blood and the interstitial fluid of the brain (blood-brain barrier) that controls the passage of molecules and cells in and out of the CNS. Several CNS diseases include leukocyte extravasation through the endothelium via two mechanistically distinct routes, the paracellular and the transcellular pathway. We established a new in vitro model of the inflamed blood-brain barrier consisting of primary cultured porcine brain capillary endothelial cells which express a tight endothelial barrier even under inflammatory conditions. By means of this specialized blood-brain barrier model we extensively studied the transmigration of neutrophils. Electron and scanning force microscopy as well as immunofluorescence imaging captured the penetrating neutrophil on the endothelial cellular body in between the junctions clearly suggesting a transcellular migration pathway. Electric cell-substrate impedance sensing and transendothelial electrical resistance measurements in combination with expression analysis of tight junction proteins demonstrate that the neutrophil-endothelial interaction does not disrupt the barrier. In conclusion, this study, based on an in vitro model of the blood-brain barrier under inflammatory conditions, evidently implicates that neutrophils preferentially migrate across the BBB via the transcellular route without impairing endothelial barrier function whereas paracellular transmigration plays only a minor role if the barrier is strongly expressed.
Collapse
Affiliation(s)
- Magdalena von Wedel-Parlow
- Institut für Biochemie, Westfälische Wilhelms-Universität Münster, Wilhelm-Klemm-Straße 2, D-48149 Münster, Germany
| | | | | | | | | | | |
Collapse
|
34
|
Wilson EH, Weninger W, Hunter CA. Trafficking of immune cells in the central nervous system. J Clin Invest 2010; 120:1368-79. [PMID: 20440079 DOI: 10.1172/jci41911] [Citation(s) in RCA: 394] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The CNS is an immune-privileged environment, yet the local control of multiple pathogens is dependent on the ability of immune cells to access and operate within this site. However, inflammation of the distinct anatomical sites (i.e., meninges, cerebrospinal fluid, and parenchyma) associated with the CNS can also be deleterious. Therefore, control of lymphocyte entry and migration within the brain is vital to regulate protective and pathological responses. In this review, several recent advances are highlighted that provide new insights into the processes that regulate leukocyte access to, and movement within, the brain.
Collapse
Affiliation(s)
- Emma H Wilson
- Division of Biomedical Sciences, University of California, Riverside, California 92521, USA.
| | | | | |
Collapse
|
35
|
Ziebell JM, Morganti-Kossmann MC. Involvement of pro- and anti-inflammatory cytokines and chemokines in the pathophysiology of traumatic brain injury. Neurotherapeutics 2010; 7:22-30. [PMID: 20129494 PMCID: PMC5084109 DOI: 10.1016/j.nurt.2009.10.016] [Citation(s) in RCA: 520] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2009] [Revised: 10/27/2009] [Accepted: 10/28/2009] [Indexed: 12/12/2022] Open
Abstract
Despite dramatic improvements in the management of traumatic brain injury (TBI), to date there is no effective treatment available to patients, and morbidity and mortality remain high. The damage to the brain occurs in two phases, the initial primary phase being the injury itself, which is irreversible and amenable only to preventive measures to minimize the extent of damage, followed by an ongoing secondary phase, which begins at the time of injury and continues in the ensuing days to weeks. This delayed phase leads to a variety of physiological, cellular, and molecular responses aimed at restoring the homeostasis of the damaged tissue, which, if not controlled, will lead to secondary insults. The development of secondary brain injury represents a window of opportunity in which pharmaceutical compounds with neuroprotective properties could be administered. To establish effective treatments for TBI victims, it is imperative that the complex molecular cascades contributing to secondary injury be fully elucidated. One pathway known to be activated in response to TBI is cellular and humoral inflammation. Neuroinflammation within the injured brain has long been considered to intensify the damage sustained following TBI. However, the accumulated findings from years of clinical and experimental research support the notion that the action of inflammation may differ in the acute and delayed phase after TBI, and that maintaining limited inflammation is essential for repair. This review addresses the role of several cytokines and chemokines following focal and diffuse TBI, as well as the controversies around the use of therapeutic anti-inflammatory treatments versus genetic deletion of cytokine expression.
Collapse
Affiliation(s)
- Jenna M. Ziebell
- grid.1002.30000000419367857National Trauma Research Institute (NTRI), The Alfred Hospital, and Department of Medicine, Monash University, 3181 Melbourne, VIC Australia
| | - Maria Cristina Morganti-Kossmann
- grid.1002.30000000419367857National Trauma Research Institute (NTRI), The Alfred Hospital, and Department of Medicine, Monash University, 3181 Melbourne, VIC Australia
| |
Collapse
|
36
|
Toledo-Pereyra LH, Lopez-Neblina F, Toledo AH. Protein Kinases in Organ Ischemia and Reperfusion. J INVEST SURG 2009; 21:215-26. [DOI: 10.1080/08941930802130149] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
37
|
Lopez-Neblina F, Toledo AH, Toledo-Pereyra LH. Molecular Biology of Apoptosis in Ischemia and Reperfusion. J INVEST SURG 2009; 18:335-50. [PMID: 16319055 DOI: 10.1080/08941930500328862] [Citation(s) in RCA: 147] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
This study reviews the current understanding of the mechanisms that mediate the complex processes involved in apoptosis secondary to ischemia and reperfusion (I/R) and is not intended as a complete literature review of apoptosis. Several biochemical reactions trigger a cascade of events, which activate caspases. These caspases exert their effect through downstream proteolysis until the final effector caspases mediate the nuclear features characteristic of apoptosis, DNA fragmentation and condensation. Within the context of ischemia, the hypoxic environment initiates the expression of several genes involved in inflammation, the immune response, and apoptosis. Many of these same genes are activated during reperfusion injury in response to radical oxygen species generation. It is plausible that inhibition of specific apoptotic pathways via inactivation or downregulation of those genes responsible for the initiation of inflammation, immune response, and apoptosis may provide promising molecular targets for ameliorating reperfusion injury in I/R-related processes. Such inhibitory mechanisms are discussed in this review. Important targets in I/R-related pathologies include the brain during stroke, the heart during myocardial infarction, and the organs during harvesting and/or storage for transplantation. In addition, we present data from our ongoing research of specific signal transduction-related elements and their role in ischemia/reperfusion injury. These data address the potential therapeutic application of anti-inflammatory and anti-ischemic compounds in the prevention of I/R damage.
Collapse
Affiliation(s)
- Fernando Lopez-Neblina
- Trauma, Surgery Research, and Molecular Biology, Borgess Research Institute, Kalamazoo, Michigan 49048, USA
| | | | | |
Collapse
|
38
|
Vilela MC, Mansur DS, Lacerda-Queiroz N, Rodrigues DH, Lima GK, Arantes RME, Kroon EG, Da Silva Campos MA, Teixeira MM, Teixeira AL. The Chemokine CCL5 Is Essential for Leukocyte Recruitment in a Model of Severe Herpes simplex Encephalitis. Ann N Y Acad Sci 2009; 1153:256-63. [DOI: 10.1111/j.1749-6632.2008.03959.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
39
|
Wunder A, Klohs J, Dirnagl U. Non-invasive visualization of CNS inflammation with nuclear and optical imaging. Neuroscience 2008; 158:1161-73. [PMID: 18983900 DOI: 10.1016/j.neuroscience.2008.10.005] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2008] [Revised: 10/01/2008] [Accepted: 10/01/2008] [Indexed: 11/28/2022]
Abstract
Inflammation is crucially involved in many diseases of the CNS. Immune cells may attack the CNS, as in multiple sclerosis, and therefore be responsible for primary damage. Immune cells may also be activated by injury to the CNS, as for example in stroke or brain trauma, secondarily enhancing lesion growth. In general, CNS inflammation involves a complex interplay of pro- and anti-inflammatory cells and molecules. The blood-brain barrier loses its integrity, plasma proteins leak into the CNS parenchyma, followed by invasion of blood-borne immune cells, and activation of resident microglial cells and astrocytes. However, inflammation not only exacerbates CNS disease, it is also indispensable in containment and resolution of tissue damage, as well as repair and regeneration. The time course and the contribution of inflammatory processes to the pathophysiology of the disease depend on several factors including the type of injury and the time point after injury, and can exhibit a high individual variability. Imaging technologies that enable specific visualization of these inflammatory processes non-invasively are therefore highly desirable. They provide powerful tools to further evaluate the contribution of specific processes to the pathophysiology of CNS disease. Moreover, these technologies may be valuable in detecting and assessing disease progression, in stratifying patients for therapy, and in monitoring therapy. Among the existing non-invasive imaging methods to visualize neuroinflammation in the CNS, we here review the current status of nuclear and optical imaging techniques, with particular emphasis on the sensitivity, specificity, as well as the limitations of these approaches.
Collapse
Affiliation(s)
- A Wunder
- Center for Stroke Research Berlin (CSB), Department of Experimental Neurology, Charité-University Medicine Berlin, Charitéplatz 1, 10117 Berlin, Germany.
| | | | | |
Collapse
|
40
|
Traffic of leukocytes in the central nervous system is associated with chemokine up-regulation in a severe model of herpes simplex encephalitis: an intravital microscopy study. Neurosci Lett 2008; 445:18-22. [PMID: 18782601 DOI: 10.1016/j.neulet.2008.08.072] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2008] [Revised: 08/26/2008] [Accepted: 08/27/2008] [Indexed: 11/22/2022]
Abstract
Herpes simplex virus type 1 (HSV-1) is a human pathogen that may cause severe encephalitis. The development of experimental models of HSV-1 encephalitis is relevant for the comprehension of the immune mechanisms involved in this infection. C57BL/6 mice were inoculated intracranially with 10(4) PFU of neurotropic HSV-1. All animals developed signs of encephalitis and died until day 6 post-infection (pi). Using intravital microscopy, we demonstrated increased leukocyte rolling and adhesion in the brain microvasculature of infected mice at days 1, 3 and 5 pi. The infection was followed by a significant increase in chemokine levels, including CCL2, CCL3, CCL5, CXCL1 and CXCL9. TNF-alpha also showed a significant increase at day 3 pi. Histological analyses demonstrated diffuse meningoencephalitis characterized mainly by mononuclear cell infiltrates. The present model of HSV-1 encephalitis exhibits high mortality in the very first days of infection. Accordingly, there were increased rolling and adhesion of leukocytes along the brain endothelium wall and a high expression of chemokines in the central nervous system. These results corroborate the role of chemokines in leukocyte recruitment following HSV-1 infection in the central nervous system.
Collapse
|
41
|
Abstract
BACKGROUND Brain death donors are frequently used for transplantation. Previous studies showed that brain death (BD) negatively affects the immunological and inflammatory status of both liver and kidney. Because the intestine is increasingly used as a donor organ and no information on effects of BD on small intestine is available we performed this study. METHODS We studied the inflammatory and apoptotic changes in donor intestine after BD induction. Brain death was induced in rats by inflation of a balloon catheter. Three groups (n=6) were compared: 1-hr BD, 4-hr BD, and sham-operated controls. RESULTS An increased polymorphonuclear cell influx in ileum, as a measure of inflammation, was observed in 1- and 4-hr BD group compared with controls. Jejunum showed a significant increase at the 4-hr BD group compared with the control group. Intercellular adhesion molecule-1, vascular cell adhesion molecule-1, E-selectin, and interleukin-6 were upregulated after 1- and 4-hr BD. Caspase-3 positive cells were found in jejunum and ileum after 4-hr BD on the top of the villi. Serum interleukin-6 was severely elevated in the 1- and 4-hr brain dead rats. CONCLUSION These data show the early occurrence of intestinal inflammation and apoptosis after BD induction. These events may ultimately have a negative influence on the outcome of intestinal transplantation.
Collapse
|
42
|
Zhang Z, Fauser U, Schluesener HJ. Dexamethasone suppresses infiltration of RhoA+ cells into early lesions of rat traumatic brain injury. Acta Neuropathol 2008; 115:335-43. [PMID: 17929039 DOI: 10.1007/s00401-007-0301-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2007] [Revised: 09/02/2007] [Accepted: 09/18/2007] [Indexed: 01/19/2023]
Abstract
Inflammatory cell infiltration is a major part of secondary tissue damage in traumatic brain injury (TBI). RhoA is an important member of Rho GTPases and is involved in leukocyte migration. Inhibition of RhoA and its downstream target, Rho-associated coiled kinase (ROCK), has been proven to promote axon regeneration and function recovery following injury in the central nervous system (CNS). Previously, we showed that dexamethasone, an immunosuppressive corticosteroid, attenuated early expression of three molecules associated with microglia/macrophages activation following TBI in rats. Here, the effects of dexamethasone on the early expression of RhoA have been investigated in brains of TBI rats by immunohistochemistry. In brains of rats treated with TBI alone, significant RhoA+ cell accumulation was observed at 18 h post-injury and continuously increased during our observed time period. The accumulated RhoA+ cells were distributed to the areas of pannecrosis and selective neuronal loss. Most accumulated RhoA+ cells were identified as active microglia/macrophages by double-labelling. Dexamethasone (1 mg/kg body weight) was intraperitoneally injected on day 0 and 2 immediately following brain injury. Numbers of RhoA+ cells were significantly reduced on day 1 and 2 following administration of dexamethasone but returned to vehicle control level on day 4. However, dexamethasone treatment did not change the proportion of RhoA+ cells. These observations suggest that dexamethasone has only a transient effect on early leukocyte recruitment.
Collapse
Affiliation(s)
- Zhiren Zhang
- Institute of Brain Research, University of Tuebingen, Calwer Str. 3, 72076 Tuebingen, Germany.
| | | | | |
Collapse
|
43
|
Scanlan BJ, Tuft B, Elfrey JE, Smith A, Zhao A, Morimoto M, Chmielinska JJ, Tejero-Taldo MI, Mak IT, Weglicki WB, Shea-Donohue T. Intestinal inflammation caused by magnesium deficiency alters basal and oxidative stress-induced intestinal function. Mol Cell Biochem 2007; 306:59-69. [PMID: 17657590 DOI: 10.1007/s11010-007-9554-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2007] [Accepted: 07/12/2007] [Indexed: 11/29/2022]
Abstract
The aim of this study was to determine the effect of magnesium deficiency on small intestinal morphology and function. Rats were assigned to 4 groups and placed on magnesium sufficient or deficient diet for 1 or 3 weeks. Infiltration of neutrophils and mucosal injury were assessed in stained sections of small intestine. Magnesium deficiency alone induced a significant increase in neutrophil infiltration and increased vascular ICAM-1 expression, in the absence of changes in mucosal injury or expression of proinflammatory mediators. Magnesium deficiency was associated with hyposecretory epithelial cell responses and vascular macromolecular leak in the small intestine and lung, which was attributed partly to reduced expression of NOS-3. To determine the effect of hypomagnesmia on the intestinal responses to a known oxidative stress, groups of rats were randomized to either sham operation or superior mesenteric artery occlusion for 10 (non-injurious) or 30 (injurious) minutes followed by a 1- or 4-hour reperfusion period. In response to mesenteric ischemia/reperfusion, deficient rats showed exaggerated PMN influx, but similar mucosal injury. Intestinal ischemia in sufficient animals induced vascular macromolecular leak in the small intestine and lung at 4 hours of reperfusion, with levels similar to those observed in untreated deficient rats. Acute magnesium repletion of deficient rats 24 h before surgery attenuated the exaggerated inflammation in deficient rats. These data show that magnesium deficiency induced a subclinical inflammation in the small intestine in the absence of mucosal injury, but with significant functional changes in local and remote organs and increased sensitivity to oxidative stress.
Collapse
Affiliation(s)
- Bradford J Scanlan
- Department of Surgery, Walter Reed Army Medical Center, Washington, DC 20307, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Koorts AM, Viljoen M. Ferritin and ferritin isoforms II: protection against uncontrolled cellular proliferation, oxidative damage and inflammatory processes. Arch Physiol Biochem 2007; 113:55-64. [PMID: 17558604 DOI: 10.1080/13813450701422575] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Ferritin is a major iron storage protein involved in the regulation of iron availability. Each ferritin molecule comprises 24 subunits. Various combinations of H-subunits and L-subunits make up the 24-subunit protein structure and these ferritin isoforms differ in their H-subunit to L-subunit ratio, as well as in their metabolic properties. Ferritin is an acute-phase protein and its expression is up-regulated in conditions such as uncontrolled cellular proliferation, in any condition marked by excessive production of toxic oxygen radicals, and by infectious and inflammatory processes. Under such conditions ferritin up-regulation is predominantly stimulated by increased reactive oxygen radical production and by cytokines. The major function of ferritin in these conditions is to reduce the bio-availability of iron in order to stem uncontrolled cellular proliferation and excessive production of reactive oxygen radicals. Ferritin is not, however, indiscriminately up-regulated in these conditions as a marked shift towards a predominance in H-subunit rich ferritins occurs. Preliminary indications are that, while the L-subunit primarily fulfils the conventional iron storage role, the H-subunit functions primarily as rapid regulator of iron availability, and perhaps indirectly as regulator of other cellular processes. It is suggested that the optimum differential expression of the two subunits differ for different cells and under different conditions and that the expression of appropriate isoferritins offers protection against uncontrolled cellular proliferation, oxidative stress and against side effects of infectious and inflammatory conditions.
Collapse
Affiliation(s)
- A M Koorts
- Department of Physiology, School of Medicine, University of Pretoria, Pretoria, South Africa.
| | | |
Collapse
|
45
|
Thomale UW, Bender M, Casalis P, Rupprecht S, Griebenow M, Neumann K, Woiciechowsky C, Unterberg AW, Stover JF. Tacrolimus depresses local immune cell infiltration but fails to reduce cortical contusion volume in brain-injured rats. Immunobiology 2007; 212:567-76. [PMID: 17678714 DOI: 10.1016/j.imbio.2007.01.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2006] [Revised: 01/15/2007] [Accepted: 01/17/2007] [Indexed: 11/24/2022]
Abstract
The immunosuppressant drug tacrolimus (FK-506) failed to show an anti-edematous effect despite suppressing pro-inflammatory cytokines in cerebrospinal fluid following focal traumatic brain injury. By questioning the role of the inflammatory response as a pharmacological target, we investigated the effects of FK-506 on immune cell infiltration in brain-injured rats. Following induction of a cortical contusion, male Sprague-Dawley rats received FK-506 or physiological saline intraperitoneally. Brains were removed at 24 h, 72 h or 7 days, respectively. Frozen brain sections (7 microm) were stained immunohistologically for markers of endothelial activation (intercellular adhesion molecule-1--ICAM-1), neutrophil infiltration (His-48), and microglial and macrophage activation (Ox-6; ED-1), respectively. Immunopositive cells were counted microscopically. Contusion volume (CV) was quantified morphometrically 7 days after trauma. Inflammatory response was confined to the ipsilateral cortex and hippocampal formation, predominating in the contusion and pericontusional cortex. Strongest ICAM-1 expression coincided with sustained granulocyte accumulation at 72h which was suppressed by FK-506. Ox-6+ cells prevailing at 72 h were also significantly reduced by FK-506. ED-1+ cells reaching highest intensity at 7 days were significantly attenuated at 72 h. Cortical CV was not influenced. FK-506 significantly decreased post-traumatic local inflammation which, however, was not associated with a reduction in cortical CV. These results question the importance of post-traumatic local immune cell infiltration in the secondary growth of a cortical contusion.
Collapse
Affiliation(s)
- Ulrich W Thomale
- Department of Neurosurgery, Charité, Medical University of Berlin, Campus Virchow Medical Center, D-13353 Berlin, Germany, and University Hospital Zürich, Division of Surgical Instensive Care Medicine, Switzerland.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Fan X, Venegas R, Fey R, van der Heyde H, Bernard MA, Lazarides E, Woods CM. An in vivo approach to structure activity relationship analysis of peptide ligands. Pharm Res 2007; 24:868-79. [PMID: 17377744 DOI: 10.1007/s11095-007-9238-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2006] [Accepted: 01/05/2007] [Indexed: 11/30/2022]
Abstract
PURPOSE The goals in this study were several-fold. First, to optimize the in vivo phage display methodology by incorporating phage pharmacokinetic properties, to isolate peptides that target the brain microvasculature, and then to build focused libraries to obtain structure activity relationship information in vivo to identify the optimal targeting motif. MATERIALS AND METHODS The blood pharmacokinetics of filamentous and T7 phage were evaluated to choose the optimal platform. A randomized peptide library with a motif CX(10)C was constructed in T7 phage and used for in vivo panning. Focused peptide libraries around each structural element of the brain-specific peptide were constructed to perform kinetic structure activity relationship (kSAR) analysis in vivo. To determine potential function, sepsis was induced in mice by LPS administration and four hours later the effect of GST-peptide on adhesion of rhodamine-labelled lymphocytes or CFDA-labelled platelets to pial microvasculature was observed by intravital microscopy. RESULTS The blood phamacokinetics of T7 was rapid (half-life of 12 min) which aids the clearance of non-specific phage. In vivo panning in brain enriched for isolates expressing the motif CAGALCY. Kinetic analysis of focused libraries built around each structural element of the peptide provided for rapid pharmacophore mapping. The computer modeling data suggested the peptide showed similarities to peptide mimetics of adhesion molecule ligands. GST-CAGALCY but not GST control protein was able to inhibit the rolling and adhesion of labeled platelets to inflamed pial vasculature. GST-CAGALCY had no effect on lymphocyte adhesion. CONCLUSIONS Incorporating normal blood phamacokinetics of T7 phage into in vivo phage display improves the ability to recover targeting peptide motifs and allows effective lead optimization by kSAR. This approach led to the isolation of a brain-specific peptide, CAGALCY, which appears to function as an effective antagonist of platelet adhesion to activated pial microvasculature.
Collapse
Affiliation(s)
- Xiaomin Fan
- AvantGen, Inc., 9924 Mesa Rim Rd, San Diego, California 92121, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Hiraga N, Adachi N, Liu K, Nagaro T, Arai T. Suppression of inflammatory cell recruitment by histamine receptor stimulation in ischemic rat brains. Eur J Pharmacol 2007; 557:236-44. [PMID: 17169356 DOI: 10.1016/j.ejphar.2006.11.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2006] [Revised: 11/06/2006] [Accepted: 11/08/2006] [Indexed: 10/23/2022]
Abstract
Inflammation is a crucial factor in the development of ischemia-induced brain injury. Since facilitation of central histaminergic activity ameliorates reperfusion injury, effects of postischemic administration of L-histidine, a precursor of histamine, and thioperamide, a histamine H3 receptor antagonist, on inflammatory cell infiltration were evaluated in a rat model of transient occlusion of the middle cerebral artery. After reperfusion for 12, 24, or 72 h following 2 h of occlusion, brain slices were immunohistochemically stained with antibodies against myeloperoxidase and CD68, which were markers of polymorphonuclear leukocytes and macrophages/microglia, respectively. After reperfusion for 12-24 h, the number of neutrophils on the ischemic side increased markedly, whereas the increase was not observed on the contralateral side. Administration of L-histidine (1000 mg/kg x 2, i.p.), immediately and 6 h after reperfusion, reduced the number of neutrophils to 52%. Simultaneous administration of thioperamide (5 mg/kg, s.c.) further decreased the number of neutrophils to 32%. Likewise, the ischemia induced increase in the number of CD68-positive cells after 24 h was suppressed by L-histidine injections. The L-histidine administration decreased the number of CD4+ T lymphocytes on both ischemic and contralateral sides after 12 h, and concurrent administration of thioperamide prolonged the effect. Although administration of mepyramine (3 nmol, i.c.v.) did not affect suppression of leukocyte infiltration, ranitidine tended to reverse the effect of L-histidine. These data suggest that enhancement of central histaminergic activity suppresses inflammatory cell recruitment after ischemic events through histamine H2 receptors, which may be a mechanism underlying the protective effect of L-histidine.
Collapse
Affiliation(s)
- Norihito Hiraga
- Department of Anesthesiology and Resuscitology, Ehime University Graduate School of Medicine, Shitsukawa, Touon-shi, Ehime 791-0295, Japan.
| | | | | | | | | |
Collapse
|
48
|
Wagner EM, Jenkins J. Effects of airway distension on leukocyte recruitment in the mouse tracheal microvasculature. J Appl Physiol (1985) 2007; 102:1528-34. [PMID: 17204569 DOI: 10.1152/japplphysiol.01054.2006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We have shown previously that excessive distention of the rat trachea during mechanical ventilation results in enhanced leukocyte recruitment to the airway (Lim LH and Wagner EM. Am J Respir Crit Care Med 168:1068-1074, 2003). The objectives of this study were to develop a mouse model of positive end-expiratory pressure (PEEP)-induced leukocyte recruitment to the airway and begin to pursue molecular mechanisms that may contribute to the in vivo observation of increased leukocyte adhesion after PEEP exposure. We studied C57BL/6 wild-type mice and mice deficient in P-selectin or intercellular adhesion molecule-1 (ICAM-1) exposed to intermittent PEEP (8 cmH(2)O) applied five times for a 1-min duration, at 10-min intervals. After the imposed ventilatory stress, during normal ventilation (0.2 ml/breath, no PEEP), leukocyte adhesion in tracheal postcapillary venules was determined using intravital microscopy. PEEP induced a time-dependent increase in leukocyte adhesion that was significantly increased between 0 and 60 min (P < 0.01). Furthermore, PEEP-induced leukocyte adhesion at 60 min was ablated in P-selectin- and ICAM-1-deficient mice. These findings demonstrate the essential nature of both P-selectin and ICAM-1 within airway postcapillary venular endothelium for leukocyte recruitment after airway distension.
Collapse
Affiliation(s)
- Elizabeth M Wagner
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland 21224, USA.
| | | |
Collapse
|
49
|
López-Neblina F, Toledo-Pereyra LH. Phosphoregulation of Signal Transduction Pathways in Ischemia and Reperfusion. J Surg Res 2006; 134:292-9. [PMID: 16519903 DOI: 10.1016/j.jss.2006.01.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2005] [Revised: 12/23/2005] [Accepted: 01/06/2006] [Indexed: 01/08/2023]
Abstract
Ischemia/reperfusion (I/R) injury triggered by pathogenic processes, such as organ transplant dysfunction, stroke, myocardial infarction, and shock, stimulate both immune and inflammatory pathways. Inflammatory cell activation and cytotoxic cytokine expression are associated with reperfusion injury. The activation of these inflammatory mediators initiates several interconnected downstream cascades regulated by phosphorylation and dephosphorylation reactions. These complex phosphorylation-dependent signal transduction pathways ultimately initiate nuclear transcription of inflammatory as well as anti-inflammatory genes to repair and assist in the recovery of damaged cells. Radical oxygen species (ROS) production, under ischemic conditions, initiates a cascade of events regulated by phosphorylation/dephosphorylation reactions and inflammatory gene expression. This is a review of the current understanding of the phosphoregulatory mechanisms that mediate the complex processes of signal transduction secondary to I/R injury. The rationale for inhibiting or activating signaling pathways as a promising molecular target for ameliorating reperfusion injury in I/R-related diseases, such as stroke, myocardial infarction, and storage for transplantation, is discussed on the basis of a new understanding of the mechanisms modulating phosphoregulatory pathways. In addition, we present part of our ongoing research in this field with phosphoregulatory signal transduction and its potential application.
Collapse
Affiliation(s)
- Fernando López-Neblina
- Trauma, Surgery Research and Molecular Biology, Borgess Research Institute, Kalamazoo, MI 49048, USA
| | | |
Collapse
|
50
|
Zhang Z, Artelt M, Burnet M, Trautmann K, Schluesener HJ. Early infiltration of CD8+ macrophages/microglia to lesions of rat traumatic brain injury. Neuroscience 2006; 141:637-644. [PMID: 16725271 DOI: 10.1016/j.neuroscience.2006.04.027] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2006] [Revised: 03/29/2006] [Accepted: 04/18/2006] [Indexed: 12/21/2022]
Abstract
Local inflammatory responses play an important role in mediating secondary tissue damage in traumatic brain injury. Characterization of leukocytic subpopulations contributing to the early infiltration of the damaged tissue might aid in further understanding of lesion development and contribute to definition of cellular targets for selective immunotherapy. In a rat traumatic brain injury model, significant CD8+ cell accumulation was observed 3 days post-injury. The CD8+ cells were strictly distributed to the pannecrotic areas and around the pannecrotic perimeter. The morphology, time course of accumulation and distribution of CD8+ cells were similar to that of reactive ED1+ and endothelial monocyte-activating polypeptide II+ microglia/macrophages, but different from W3/13+ T cells. Further double-labeling experiments confirmed that the major cellular sources of CD8 were reactive macrophages/microglia. Both the location of these CD8+ macrophages/microglia to the border of the pannecrosis and their co-expression of endothelial monocyte-activating polypeptide II and P2X4 receptor suggest they might have a central role in lesion development and might thus be candidates for development of immunotherapeutic, anti-inflammatory strategies.
Collapse
Affiliation(s)
- Z Zhang
- Institute of Brain Research, University of Tuebingen, Calwer Str. 3, D-72076 Tuebingen, Germany.
| | - M Artelt
- Synovo GmbH, Paul-Ehrlich-Str. 15, D-72076 Tuebingen, Germany
| | - M Burnet
- Synovo GmbH, Paul-Ehrlich-Str. 15, D-72076 Tuebingen, Germany
| | - K Trautmann
- Institute of Brain Research, University of Tuebingen, Calwer Str. 3, D-72076 Tuebingen, Germany
| | - H J Schluesener
- Institute of Brain Research, University of Tuebingen, Calwer Str. 3, D-72076 Tuebingen, Germany
| |
Collapse
|