1
|
Dwyer MKR, Amelinez-Robles N, Polsfuss I, Herbert K, Kim C, Varghese N, Parry TJ, Buller B, Verdoorn TA, Billing CB, Morrison B. NTS-105 decreased cell death and preserved long-term potentiation in an in vitro model of moderate traumatic brain injury. Exp Neurol 2024; 371:114608. [PMID: 37949202 DOI: 10.1016/j.expneurol.2023.114608] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/27/2023] [Accepted: 11/05/2023] [Indexed: 11/12/2023]
Abstract
Traumatic brain injury (TBI) is a major cause of hospitalization and death. To mitigate these human costs, the search for effective drugs to treat TBI continues. In the current study, we evaluated the efficacy of the novel neurosteroid, NTS-105, to reduce post-traumatic pathobiology in an in vitro model of moderate TBI that utilizes an organotypic hippocampal slice culture. NTS-105 inhibited activation of the androgen receptor and the mineralocorticoid receptor, partially activated the progesterone B receptor and was not active at the glucocorticoid receptor. Treatment with NTS-105 starting one hour after injury decreased post-traumatic cell death in a dose-dependent manner, with 10 nM NTS-105 being most effective. Post-traumatic administration of 10 nM NTS-105 also prevented deficits in long-term potentiation (LTP) without adversely affecting neuronal activity in naïve cultures. We propose that the high potency pleiotropic action of NTS-105 beneficial effects at multiple receptors (e.g. androgen, mineralocorticoid and progesterone) provides significant mechanistic advantages over native neurosteroids such as progesterone, which lacked clinical success for the treatment of TBI. Our results suggest that this pleiotropic pharmacology may be a promising strategy for the effective treatment of TBI, and future studies should test its efficacy in pre-clinical animal models of TBI.
Collapse
Affiliation(s)
- Mary Kate R Dwyer
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States of America
| | - Nicolas Amelinez-Robles
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States of America
| | - Isabella Polsfuss
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States of America
| | - Keondre Herbert
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States of America
| | - Carolyn Kim
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States of America
| | - Nevin Varghese
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States of America
| | - Tom J Parry
- NeuroTrauma Sciences, LLC, Alpharetta, GA 30009, United States of America
| | - Benjamin Buller
- NeuroTrauma Sciences, LLC, Alpharetta, GA 30009, United States of America
| | - Todd A Verdoorn
- NeuroTrauma Sciences, LLC, Alpharetta, GA 30009, United States of America
| | - Clare B Billing
- BioPharmaWorks, LLC, Groton, CT 06340, United States of America
| | - Barclay Morrison
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States of America.
| |
Collapse
|
2
|
Dang Y, Wang T. Research Progress on the Immune-Inflammatory Mechanisms of Posttraumatic Epilepsy. Cell Mol Neurobiol 2023; 43:4059-4069. [PMID: 37889439 DOI: 10.1007/s10571-023-01429-2] [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: 07/20/2023] [Accepted: 10/17/2023] [Indexed: 10/28/2023]
Abstract
Posttraumatic epilepsy (PTE) is a severe complication arising from a traumatic brain injury caused by various violent actions on the brain. The underlying mechanisms for the pathogenesis of PTE are complex and have not been fully defined. Approximately, one-third of patients with PTE are resistant to antiepileptic therapy. Recent research evidence has shown that neuroinflammation is critical in the development of PTE. This article reviews the immune-inflammatory mechanisms regarding microglial activation, astrocyte proliferation, inflammatory signaling pathways, chronic neuroinflammation, and intestinal flora. These mechanisms offer novel insights into the pathophysiological mechanisms of PTE and have groundbreaking implications in the prevention and treatment of PTE. Immunoinflammatory cross-talk between glial cells and gut microbiota in posttraumatic epilepsy. This graphical abstract depicts the roles of microglia and astrocytes in posttraumatic epilepsy, highlighting the influence of the gut microbiota on their function. TBI traumatic brain injury, AQP4 aquaporin-4, Kir4.1 inward rectifying K channels.
Collapse
Affiliation(s)
- Yangbin Dang
- Department of Neurology, Epilepsy Center, Lanzhou University Second Hospital, No. 82 Cuiyingmen, Lanzhou, 730000, Gansu, China
| | - Tiancheng Wang
- Department of Neurology, Epilepsy Center, Lanzhou University Second Hospital, No. 82 Cuiyingmen, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
3
|
El Sayed R, Shankar KM, Mankame AR, Cox CS. Innovations in cell therapy in pediatric diseases: a narrative review. Transl Pediatr 2023; 12:1239-1257. [PMID: 37427072 PMCID: PMC10326759 DOI: 10.21037/tp-23-92] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 05/25/2023] [Indexed: 07/11/2023] Open
Abstract
Background and Objective Stem cell therapy is a regenerative medicine modality that has the potential to decrease morbidity and mortality by promoting tissue regeneration or modulating the inflammatory response. An increase in the number of clinical trials investigating the efficacy and safety of stem cell therapy in pediatric diseases has led to advancements in this field. Currently, multiple sources and types of stem cells have been utilized in the treatment of pediatric diseases. This review aims to inform researchers and clinicians about preclinical and clinical stem cell therapy trials in pediatric patients. We discuss the different types of stem cells and the wide spectrum of stem cell therapy trials for pediatric diseases, with an emphasis on the outcomes and advancements in the field. Methods PubMed and clinicaltrials.gov databases were searched on October 28, 2022 using the following Medical Subject Headings (MeSH) terms "stem cell" or "stem cell therapy" with an age filter <18 years. Our search was limited to publications published between 2000 and 2022. Key Content and Findings Diverse sources of stem cells have different properties and mechanisms of action, which allow tailored application of stem cells according to the pathophysiology of the disease. Advancements in stem cell therapies for pediatric diseases have led to improvements in clinical outcomes in some pediatric diseases or in quality of life, such therapies represent a potential alternative to the current treatment modalities. Conclusions Stem cell therapy in pediatric diseases has shown promising results and outcomes. However, further studies focusing on the implementation and optimal treatment timeframe are needed. An increase in preclinical and clinical trials of stem cell therapy targeting pediatric patients is required to advance our therapeutic applications.
Collapse
Affiliation(s)
- Razan El Sayed
- Department of Pediatric Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Center for Translational Injury Research, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Department of Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Karan Michael Shankar
- Department of Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Atharwa Rajan Mankame
- Department of Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Charles S. Cox
- Department of Pediatric Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Center for Translational Injury Research, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Department of Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| |
Collapse
|
4
|
Bolden CT, Olson SD, Cox CS. A decade of blood-brain barrier permeability assays: Revisiting old traumatic brain injury rat data for new insights and experimental design. Microvasc Res 2023; 145:104453. [PMID: 36356686 PMCID: PMC9712264 DOI: 10.1016/j.mvr.2022.104453] [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: 06/01/2022] [Revised: 10/01/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022]
Abstract
Increased microvascular permeability at the level of the blood-brain barrier (BBB) often leads to vasogenic brain edema following traumatic brain injury (TBI). These pathologic conditions compromise the integrity of the neurovascular unit resulting in severe brain dysfunction. To quantify this permeability and assess ionic equillibrium, preclinical researchers have relied on the use of various molecular weight permeable dyes such as Evans Blue that normally cannot enter the brain parenchyma under homeostatic conditions. Evans Blue, the most cited of the molecular weight dyes, has reported reproducibility issues because of harsh extraction processes, suboptimal detection via absorbance, and wide excitation fluorescence spectra associated with the dye. Our laboratory group transitioned to Alexa Fluor 680, a far-red dye with improved sensitivity compared to Evans Blue and thus improved reproducibility to alleviate this issue. To evaluate our reproducibility and increase the rigor of our experimental design, we retrospectively analyzed our controlled cortical impact (CCI) experiments over the past 10 years to evaluate effect size with larger samples and potential sources of variability. All of our BBB permeability experiments were performed with Male, Sprague Dawley rats weighing between 225 and 300 g. Historically, Sprague Dawleys were randomly divided into treatment groups: SHAM, CCI, and a stem cell-based treatment from years 2007-2020. The assessment of microvascular hyperpermeability were evaluated by comparing the mean at minimum threshold, area at 1 k-2 k, and intensity density obtained from Alexa Fluor 680 permeability data. Studies utilizing Evans Blue were further compared by tip depth, diameter size, and the hemisphere of injury. Statistical evaluation utilizing the G Power software analysis did not yield a significant difference in sample size comparing experimental groups for Evans Blue and Alexa Fluor 680 analyzed brain tissue. Our analysis also demonstrated a trend in that recent studies (years 2018-2020) have yielded more compact sample sizes between experimental groups in Alexa Fluor 680 analyzed rats. This retrospective study further revealed that Alexa Fluor 680 image analysis provides greater sensitivity to BBB permeability following TBI in comparison to Evans Blue. Significant differences in sample size were not detected between Evans Blue and Alexa Fluor 680; there were significant differences found throughout year to year analysis at the lower range of thresholds. SUMMARY STATEMENT: This work provides a comparative analysis of BBB permeability assay techniques after CCI model of injury in rats.
Collapse
Affiliation(s)
- Chris T Bolden
- Department of Pediatric Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Center for Translational Injury Research, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Scott D Olson
- Department of Pediatric Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Charles S Cox
- Department of Pediatric Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
| |
Collapse
|
5
|
Gao Z, Pang Z, Lei G, Chen Y, Cai Z, Zhu S, Lin W, Qiu Z, Wang Y, Shen Y, Xu W. Crossing nerve transfer drives sensory input-dependent plasticity for motor recovery after brain injury. SCIENCE ADVANCES 2022; 8:eabn5899. [PMID: 36044580 PMCID: PMC9432844 DOI: 10.1126/sciadv.abn5899] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 07/18/2022] [Indexed: 06/15/2023]
Abstract
Restoring limb movements after central nervous system injury remains a substantial challenge. Recent studies proved that crossing nerve transfer surgery could rebuild physiological connectivity between the contralesional cortex and the paralyzed arm to compensate for the lost function after brain injury. However, the neural mechanism by which this surgery mediates motor recovery remains still unclear. Here, using a clinical mouse model, we showed that this surgery can restore skilled forelimb function in adult mice with unilateral cortical lesion by inducing cortical remapping and promoting corticospinal tract sprouting. After reestablishing the ipsilateral descending pathway, resecting of the artificially rebuilt peripheral nerve did not affect motor improvements. Furthermore, retaining the sensory afferent, but not the motor efferent, of the transferred nerve was sufficient for inducing brain remapping and facilitating motor restoration. Thus, our results demonstrate that surgically rebuilt sensory input triggers neural plasticity for accelerating motor recovery, which provides an approach for treating central nervous system injuries.
Collapse
Affiliation(s)
- Zhengrun Gao
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhen Pang
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Gaowei Lei
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yiming Chen
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Zeyu Cai
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Shuai Zhu
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Weishan Lin
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Zilong Qiu
- The National Clinical Research Center for Aging and Medicine, Fudan University, Shanghai, China
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yizheng Wang
- The National Clinical Research Center for Aging and Medicine, Fudan University, Shanghai, China
| | - Yundong Shen
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China
- The National Clinical Research Center for Aging and Medicine, Fudan University, Shanghai, China
- Department of Hand and Upper Extremity Surgery, Jing‘an District Central Hospital, Fudan University, Shanghai, China
| | - Wendong Xu
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China
- The National Clinical Research Center for Aging and Medicine, Fudan University, Shanghai, China
- Department of Hand and Upper Extremity Surgery, Jing‘an District Central Hospital, Fudan University, Shanghai, China
- Institutes of Brain Science, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center of Brain Science, Fudan University, Shanghai, China
- Co-innovation Center of Neuroregeneration, Nantong University, 226000 Nantong, China
- Research Unit of Synergistic Reconstruction of Upper and Lower Limbs After Brain Injury, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
6
|
Klein L, Ophelders DR, van den Hove D, Damoiseaux M, Rutten BP, Reutelingsperger CP, Schurgers LJ, Wolfs TG. Prenatal administration of multipotent adult progenitor cells modulates the systemic and cerebral immune response in an ovine model of chorioamnionitis. Brain Behav Immun Health 2022; 23:100458. [PMID: 35647567 PMCID: PMC9136278 DOI: 10.1016/j.bbih.2022.100458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/17/2022] [Accepted: 03/31/2022] [Indexed: 11/30/2022] Open
Abstract
Systemic and cerebral inflammation following antenatal infection (e.g. chorioamnionitis) and dysregulation of the blood brain barrier (BBB) are major risk factors for abnormal neonatal brain development. Administration of multipotent adult progenitor cells (MAPCs) represents an interesting pharmacological strategy as modulator of the peripheral and cerebral immune response and protector of BBB integrity. We studied the immunomodulatory and protective cerebrovascular potential of prenatally administered MAPCs in a preclinical ovine model for antenatal inflammation. Ovine fetuses were intra-amniotically (i.a.) exposed to lipopolysaccharide (LPS) or saline at gestational day 125, followed by the intravenous administration of 1*107 MAPCs or saline at gestational day 127. Circulating inflammation markers were measured. Fetal brains were examined immuno-histochemically post-mortem at gestational day 132. Fetal plasma IL-6 levels were elevated significantly 24 h after LPS administration. In utero systemic MAPC treatment after LPS exposure increased Annexin A1 (ANXA1) expression in the cerebrovascular endothelium, indicating enforcement of BBB integrity, and increased the number of leukocytes at brain barriers throughout the brain. Further characterisation of brain barrier-associated leukocytes showed that monocyte/choroid plexus macrophage (IBA-1+/CD206+) and neutrophil (MPO+) populations predominantly contributed to the LPS-MAPC-induced increase of CD45+cells. In the choroid plexus, the percentage of leukocytes expressing the proresolving mediator ANXA1 tended to be decreased after LPS-induced antenatal inflammation, an effect reversed by systemic MAPC treatment. Accordingly, expression levels of ANXA1 per leukocyte were decreased after LPS and restored after subsequent MAPC treatment. Increased expression of ANXA1 by the cerebrovasculature and immune cells at brain barriers following MAPC treatment in an infectious setting indicate a MAPC driven early defence mechanism to protect the neonatal brain against infection-driven inflammation and potential additional pro-inflammatory insults in the neonatal period. MAPCs administered systemically enhance the brain directed immune response in an inflammation dependent manner in preterm fetuses. Annexin A1 expression is increased in cerebrovasculature and immune cells at brain barriers when MAPCs were i.v. administered in the infectious setting. MAPCs potentially protect the neonatal brain by enforcing the blood brain barrier and modulating inflammation.
Collapse
Affiliation(s)
- Luise Klein
- School for Oncology and Reproduction (GROW), Maastricht University, Maastricht, the Netherlands
- Department of Pediatrics, Maastricht University, Maastricht, the Netherlands
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, the Netherlands
| | - Daan R.M.G. Ophelders
- School for Oncology and Reproduction (GROW), Maastricht University, Maastricht, the Netherlands
- Department of Pediatrics, Maastricht University, Maastricht, the Netherlands
| | - Daniel van den Hove
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, the Netherlands
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany
- Department of Psychiatry and Neuropsychology, European Graduate School of Neuroscience (EURON), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, the Netherlands
| | - Maurits Damoiseaux
- School for Oncology and Reproduction (GROW), Maastricht University, Maastricht, the Netherlands
- Department of Pediatrics, Maastricht University, Maastricht, the Netherlands
| | - Bart P.F. Rutten
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, the Netherlands
- Department of Psychiatry and Neuropsychology, European Graduate School of Neuroscience (EURON), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, the Netherlands
| | - Chris P.M. Reutelingsperger
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands
| | - Leon J. Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands
| | - Tim G.A.M. Wolfs
- School for Oncology and Reproduction (GROW), Maastricht University, Maastricht, the Netherlands
- Department of Pediatrics, Maastricht University, Maastricht, the Netherlands
- Corresponding author. School for Oncology and Reproduction (GROW), Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
7
|
Grade S, Thomas J, Zarb Y, Thorwirth M, Conzelmann KK, Hauck SM, Götz M. Brain injury environment critically influences the connectivity of transplanted neurons. SCIENCE ADVANCES 2022; 8:eabg9445. [PMID: 35687687 PMCID: PMC9187233 DOI: 10.1126/sciadv.abg9445] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Cell transplantation is a promising approach for the reconstruction of neuronal circuits after brain damage. Transplanted neurons integrate with remarkable specificity into circuitries of the mouse cerebral cortex affected by neuronal ablation. However, it remains unclear how neurons perform in a local environment undergoing reactive gliosis, inflammation, macrophage infiltration, and scar formation, as in traumatic brain injury (TBI). To elucidate this, we transplanted cells from the embryonic mouse cerebral cortex into TBI-injured, inflamed-only, or intact cortex of adult mice. Brain-wide quantitative monosynaptic rabies virus (RABV) tracing unraveled graft inputs from correct regions across the brain in all conditions, with pronounced quantitative differences: scarce in intact and inflamed brain versus exuberant after TBI. In the latter, the initial overshoot is followed by pruning, with only a few input neurons persisting at 3 months. Proteomic profiling identifies candidate molecules for regulation of the synaptic yield, a pivotal parameter to tailor for functional restoration of neuronal circuits.
Collapse
Affiliation(s)
- Sofia Grade
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians University Munich, 82152 Planegg-Martinsried, Germany
- Institute of Stem Cell Research, Helmholtz Center Munich, German Center for Environmental Health, 82152 Planegg-Martinsried, Germany
- Corresponding author. (S.G.); (S.M.H.); (M.G.)
| | - Judith Thomas
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians University Munich, 82152 Planegg-Martinsried, Germany
- Institute of Stem Cell Research, Helmholtz Center Munich, German Center for Environmental Health, 82152 Planegg-Martinsried, Germany
- Graduate School of Systemic Neuroscience, Ludwig-Maximilians University Munich, 82152 Planegg-Martinsried, Germany
| | - Yvette Zarb
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians University Munich, 82152 Planegg-Martinsried, Germany
- Institute of Stem Cell Research, Helmholtz Center Munich, German Center for Environmental Health, 82152 Planegg-Martinsried, Germany
| | - Manja Thorwirth
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians University Munich, 82152 Planegg-Martinsried, Germany
- Institute of Stem Cell Research, Helmholtz Center Munich, German Center for Environmental Health, 82152 Planegg-Martinsried, Germany
| | - Karl-Klaus Conzelmann
- Max von Pettenkofer Institute Virology, Medical Faculty and Gene Center, Ludwig-Maximilians University Munich, 81377 Munich, Germany
| | - Stefanie M. Hauck
- Research Unit Protein Science and Metabolomics and Proteomics Core, Helmholtz Center Munich, German Center for Environmental Health, 85764 Neuherberg, Germany
- Corresponding author. (S.G.); (S.M.H.); (M.G.)
| | - Magdalena Götz
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians University Munich, 82152 Planegg-Martinsried, Germany
- Institute of Stem Cell Research, Helmholtz Center Munich, German Center for Environmental Health, 82152 Planegg-Martinsried, Germany
- SYNERGY, Excellence Cluster of Systems Neurology, Biomedical Center, Ludwig-Maximilians University Munich, 82152 Planegg-Martinsried, Germany
- Corresponding author. (S.G.); (S.M.H.); (M.G.)
| |
Collapse
|
8
|
Cox CS, Juranek J, Kosmach S, Pedroza C, Thakur N, Dempsey A, Rennie K, Scott MC, Jackson M, Kumar A, Aertker B, Caplan H, Triolo F, Savitz SI. Autologous cellular therapy for cerebral palsy: a randomized, crossover trial. Brain Commun 2022; 4:fcac131. [PMID: 35702731 PMCID: PMC9188321 DOI: 10.1093/braincomms/fcac131] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 02/24/2022] [Accepted: 05/17/2022] [Indexed: 11/14/2022] Open
Abstract
We examined an autologous mononuclear-cell-therapy-based approach to treat cerebral palsy using autologous umbilical cord blood or bone-marrow-derived mononuclear cells. The primary objective was to determine if autologous cells are safe to administer in children with cerebral palsy. The secondary objectives were to determine if there was improvement in motor function of patients 12 months after infusion using the Gross Motor Function Measure and to evaluate impact of treatment on corticospinal tract microstructure as determined by radial diffusivity measurement. This Phase 1/2a trial was a randomized, blinded, placebo-controlled, crossover study in children aged 2-10 years of age with cerebral palsy enrolled between November 2013 and November 2016. Participants were randomized to 2:1 treatment:placebo. Treatment was either autologous bone-marrow-derived mononuclear cells or autologous umbilical cord blood. All participants who enrolled and completed their baseline visit planned to return for follow-up visits at 6 months, 12 months and 24 months after the baseline visit. At the 12-month post-treatment visit, participants who originally received the placebo received either bone-marrow-derived mononuclear cell or umbilical cord blood treatment. Twenty participants were included; 7 initially randomized to placebo, and 13 randomized to treatment. Five participants randomized to placebo received bone-marrow-derived mononuclear cells, and 2 received umbilical cord blood at the 12-month visit. None of the participants experienced adverse events related to the stem cell infusion. Cell infusion at the doses used in our study did not dramatically alter motor function. We observed concordant bilateral changes in radial diffusivity in 10 of 15 cases where each corticospinal tract could be reconstructed in each hemisphere. In 60% of these cases (6/10), concordant decreases in bilateral corticospinal tract radial diffusivity occurred post-treatment. In addition, 100% of unilateral corticospinal tract cases (3/3) exhibited decreased corticospinal tract radial diffusivity post-treatment. In our discordant cases (n = 5), directionality of changes in corticospinal tract radial diffusivity appeared to coincide with handedness. There was a significant improvement in corticospinal tract radial diffusivity that appears related to handedness. Connectivity strength increased in either or both pathways (corticio-striatal and thalamo-cortical) in each participant at 12 months post-treatment. These data suggest that both stem cell infusions are safe. There may be an improvement in myelination in some groups of patients that correlate with small improvements in the Gross Motor Function Measure scales. A larger autologous cord blood trial is impractical at current rates of blood banking. Either increased private banking or matched units would be required to perform a larger-scale trial.
Collapse
Affiliation(s)
- Charles S. Cox
- Department of Pediatric Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Program in Pediatric Regenerative Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Jenifer Juranek
- Department of Pediatric Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Program in Pediatric Regenerative Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Steven Kosmach
- Department of Pediatric Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Claudia Pedroza
- Department of Pediatrics, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Nivedita Thakur
- Department of Pediatrics, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Allison Dempsey
- Department of Pediatrics, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Kimberly Rennie
- Department of Pediatrics, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Department of Neuropsychology, NeuroBehavioral Health, Milwaukee, WI, USA
| | - Michael C. Scott
- Department of Pediatric Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Margaret Jackson
- Department of Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Akshita Kumar
- Department of Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Benjamin Aertker
- Department of Neurology, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Henry Caplan
- Department of Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Fabio Triolo
- Department of Pediatric Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Program in Pediatric Regenerative Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Sean I. Savitz
- Department of Neurology, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| |
Collapse
|
9
|
Gottlieb A, Toledano-Furman N, Prabhakara KS, Kumar A, Caplan HW, Bedi S, Cox CS, Olson SD. Time dependent analysis of rat microglial surface markers in traumatic brain injury reveals dynamics of distinct cell subpopulations. Sci Rep 2022; 12:6289. [PMID: 35428862 PMCID: PMC9012748 DOI: 10.1038/s41598-022-10419-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 04/07/2022] [Indexed: 12/15/2022] Open
Abstract
Traumatic brain injury (TBI) results in a cascade of cellular responses, which produce neuroinflammation, partly due to the activation of microglia. Accurate identification of microglial populations is key to understanding therapeutic approaches that modify microglial responses to TBI and improve long-term outcome measures. Notably, previous studies often utilized an outdated convention to describe microglial phenotypes. We conducted a temporal analysis of the response to controlled cortical impact (CCI) in rat microglia between ipsilateral and contralateral hemispheres across seven time points, identified microglia through expression of activation markers including CD45, CD11b/c, and p2y12 receptor and evaluated their activation state using additional markers of CD32, CD86, RT1B, CD200R, and CD163. We identified unique sub-populations of microglial cells that express individual or combination of activation markers across time points. We further portrayed how the size of these sub-populations changes through time, corresponding to stages in TBI response. We described longitudinal changes in microglial population after CCI in two different locations using activation markers, showing clear separation into cellular sub-populations that feature different temporal patterns of markers after injury. These changes may aid in understanding the symptomatic progression following TBI and help define microglial subpopulations beyond the outdated M1/M2 paradigm.
Collapse
Affiliation(s)
- Assaf Gottlieb
- Center for Precision Health, School of Biomedical Informatics, University of Texas Health Science Center, Houston, TX, 77030, USA.
| | - Naama Toledano-Furman
- Department of Pediatric Surgery, McGovern School of Medicine, University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX, 77030, USA
| | - Karthik S Prabhakara
- Department of Pediatric Surgery, McGovern School of Medicine, University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX, 77030, USA
| | - Akshita Kumar
- Department of Pediatric Surgery, McGovern School of Medicine, University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX, 77030, USA
| | - Henry W Caplan
- Department of Pediatric Surgery, McGovern School of Medicine, University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX, 77030, USA
| | - Supinder Bedi
- Department of Pediatric Surgery, McGovern School of Medicine, University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX, 77030, USA
| | - Charles S Cox
- Department of Pediatric Surgery, McGovern School of Medicine, University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX, 77030, USA
| | - Scott D Olson
- Department of Pediatric Surgery, McGovern School of Medicine, University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX, 77030, USA.
| |
Collapse
|
10
|
Scott MC, Prabhakara KS, Walters AJ, Olson SD, Cox CS. Determining Sex-Based Differences in Inflammatory Response in an Experimental Traumatic Brain Injury Model. Front Immunol 2022; 13:753570. [PMID: 35222368 PMCID: PMC8864286 DOI: 10.3389/fimmu.2022.753570] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction Traumatic brain injury is a leading cause of injury-related death and morbidity. Multiple clinical and pre-clinical studies have reported various results regarding sex-based differences in TBI. Our accepted rodent model of traumatic brain injury was used to identify sex-based differences in the pathological features of TBI. Methods Male and female Sprague-Dawley rats were subjected to either controlled-cortical impact (CCI) or sham injury; brain tissue was harvested at different time intervals depending on the specific study. Blood-brain barrier (BBB) analysis was performed using infrared imaging to measure fluorescence dye extravasation. Microglia and splenocytes were characterized with traditional flow cytometry; microglia markers such as CD45, P2Y12, CD32, and CD163 were analyzed with t-distributed stochastic neighbor embedding (t-SNE). Flow cytometry was used to study tissue cytokine levels, and supplemented with ELISAs of TNF-⍺, IL-17, and IL-1β of the ipsilateral hemisphere tissue. Results CCI groups of both sexes recorded a higher BBB permeability at 72 hours post-injury than their respective sham groups. There was significant difference in the integrated density value of BBB permeability between the male CCI group and the female CCI group (female CCI mean = 3.08 x 108 ± 2.83 x 107, male CCI mean = 2.20 x 108 ± 4.05 x 106, p = 0.0210), but otherwise no differences were observed. Traditional flow cytometry did not distinguish any sex-based difference in regards to splenocyte cell population after CCI. t-SNE did not reveal any significant difference between the male and female injury groups in the activation of microglia. Cytokine analysis after injury by flow cytometry and ELISA was limited in differences at the time point of 6 hours post-injury. Conclusion In our rodent model of traumatic brain injury, sex-based differences in pathology and neuroinflammation at specified time points are limited, and only noted in one specific analysis of BBB permeability.
Collapse
Affiliation(s)
- Michael C. Scott
- Department of Pediatric Surgery, University of Texas Health Science Center at Houston, Houston, TX, United States
| | | | | | - Scott D. Olson
- Department of Pediatric Surgery, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Charles S. Cox
- Department of Pediatric Surgery, University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
11
|
Pavic G, Petzsch P, Jansen R, Raba K, Rychlik N, Simiantonakis I, Küry P, Göttle P, Köhrer K, Hartung HP, Meuth SG, Jander S, Gliem M. Microglia contributes to remyelination in cerebral but not spinal cord ischemia. Glia 2021; 69:2739-2751. [PMID: 34390590 DOI: 10.1002/glia.24068] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/02/2021] [Accepted: 07/07/2021] [Indexed: 11/07/2022]
Abstract
Inflammation after injury of the central nervous system (CNS) is increasingly viewed as a therapeutic target. However, comparative studies in different CNS compartments are sparse. To date only few studies based on immunohistochemical data and all referring to mechanical injury have directly compared inflammation in different CNS compartments. These studies revealed that inflammation is more pronounced in spinal cord than in brain. Therefore, it is unclear whether concepts and treatments established in the cerebral cortex can be transferred to spinal cord lesions and vice versa or whether immunological treatments must be adapted to different CNS compartments. By use of transcriptomic and flow cytometry analysis of equally sized photothrombotically induced lesions in the cerebral cortex and the spinal cord, we could document an overall comparable inflammatory reaction and repair activity in brain and spinal cord between day 1 and day 7 after ischemia. However, remyelination was increased after cerebral versus spinal cord ischemia which is in line with increased remyelination in gray matter in previous analyses and was accompanied by microglia dominated inflammation opposed to monocytes/macrophages dominated inflammation after spinal cord ischemia. Interestingly remyelination could be reduced by microglia and not hematogenous macrophage depletion. Our results show that despite different cellular composition of the postischemic infiltrate the inflammatory response in cerebral cortex and spinal cord are comparable between day 1 and day 7. A striking difference was higher remyelination capacity in the cerebral cortex, which seems to be supported by microglia dominance.
Collapse
Affiliation(s)
- Goran Pavic
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Patrick Petzsch
- Biological and Medical Research Center (BMFZ), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Robin Jansen
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Katharina Raba
- Institute for Transplantation Diagnostics and Cell Therapeutics, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Nicole Rychlik
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | | | - Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Peter Göttle
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Karl Köhrer
- Biological and Medical Research Center (BMFZ), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Sven G Meuth
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Sebastian Jander
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Michael Gliem
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
12
|
Multipotent adult progenitor cells induce regulatory T cells and promote their suppressive phenotype via TGFβ and monocyte-dependent mechanisms. Sci Rep 2021; 11:13549. [PMID: 34193955 PMCID: PMC8245558 DOI: 10.1038/s41598-021-93025-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 06/17/2021] [Indexed: 02/08/2023] Open
Abstract
Dysregulation of the immune system can initiate chronic inflammatory responses that exacerbate disease pathology. Multipotent adult progenitor cells (MAPC cells), an adult adherent bone-marrow derived stromal cell, have been observed to promote the resolution of uncontrolled inflammatory responses in a variety of clinical conditions including acute ischemic stroke, acute myocardial infarction (AMI), graft vs host disease (GvHD), and acute respiratory distress syndrome (ARDS). One of the proposed mechanisms by which MAPC cells modulate immune responses is via the induction of regulatory T cells (Tregs), however, the mechanism(s) involved remains to be fully elucidated. Herein, we demonstrate that, in an in vitro setting, MAPC cells increase Treg frequencies by promoting Treg proliferation and CD4+ T cell differentiation into Tregs. Moreover, MAPC cell-induced Tregs (miTregs) have a more suppressive phenotype characterized by increased expression of CTLA-4, HLA-DR, and PD-L1 and T cell suppression capacity. MAPC cells also promoted Treg activation by inducing CD45RA+ CD45RO+ transitional Tregs. Additionally, we identify transforming growth factor beta (TGFβ) as an essential factor for Treg induction secreted by MAPC cells. Furthermore, inhibition of indoleamine 2, 3-dioxygenase (IDO) resulted in decreased Treg induction by MAPC cells demonstrating IDO involvement. Our studies also show that CD14+ monocytes play a critical role in Treg induction by MAPC cells. Our study describes MAPC cell dependent Treg phenotypic changes and provides evidence of potential mechanisms by which MAPC cells promote Treg differentiation.
Collapse
|
13
|
Scott MC, Bedi SS, Olson SD, Sears CM, Cox CS. Microglia as therapeutic targets after neurological injury: strategy for cell therapy. Expert Opin Ther Targets 2021; 25:365-380. [PMID: 34029505 DOI: 10.1080/14728222.2021.1934447] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Microglia is the resident tissue macrophages of the central nervous system. Prolonged microglial activation often occurs after traumatic brain injury and is associated with deteriorating neurocognitive outcomes. Resolution of microglial activation is associated with limited tissue loss and improved neurocognitive outcomes. Limiting the prolonged pro-inflammatory response and the associated secondary tissue injury provides the rationale and scientific premise for considering microglia as a therapeutic target. AREAS COVERED In this review, we discuss markers of microglial activation, such as immunophenotype and microglial response to injury, including cytokine/chemokine release, free radical formation, morphology, phagocytosis, and metabolic shifts. We compare the origin and role in neuroinflammation of microglia and monocytes/macrophages. We review potential therapeutic targets to shift microglial polarization. Finally, we review the effect of cell therapy on microglia. EXPERT OPINION Dysregulated microglial activation after neurologic injury, such as traumatic brain injury, can worsen tissue damage and functional outcomes. There are potential targets in microglia to attenuate this activation, such as proteins and molecules that regulate microglia polarization. Cellular therapeutics that limit, but do not eliminate, the inflammatory response have improved outcomes in animal models by reducing pro-inflammatory microglial activation via secondary signaling. These findings have been replicated in early phase clinical trials.
Collapse
Affiliation(s)
- M Collins Scott
- Department of Pediatric Surgery, University of Texas Health Science Center at Houston (Uthealth), USA
| | - Supinder S Bedi
- Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, Texas, USA
| | - Scott D Olson
- Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, Texas, USA
| | - Candice M Sears
- Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, Texas, USA
| | - Charles S Cox
- Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, Texas, USA
| |
Collapse
|
14
|
Microglia: A Potential Drug Target for Traumatic Axonal Injury. Neural Plast 2021; 2021:5554824. [PMID: 34093701 PMCID: PMC8163545 DOI: 10.1155/2021/5554824] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/06/2021] [Accepted: 04/26/2021] [Indexed: 12/20/2022] Open
Abstract
Traumatic axonal injury (TAI) is a major cause of death and disability among patients with severe traumatic brain injury (TBI); however, no effective therapies have been developed to treat this disorder. Neuroinflammation accompanying microglial activation after TBI is likely to be an important factor in TAI. In this review, we summarize the current research in this field, and recent studies suggest that microglial activation plays an important role in TAI development. We discuss several drugs and therapies that may aid TAI recovery by modulating the microglial phenotype following TBI. Based on the findings of recent studies, we conclude that the promotion of active microglia to the M2 phenotype is a potential drug target for the treatment of TAI.
Collapse
|
15
|
Demir S, Erturk A, Gunal YD, Ozmen I, Zengin M, Yildiz D, Karaoz E, Karahan S, Senel E. Contribution of Bone Marrow-Derived Mesenchymal Stem Cells to Healing of Pulmonary Contusion-Created Rats. J Surg Res 2021; 261:205-214. [PMID: 33450629 DOI: 10.1016/j.jss.2020.12.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 11/28/2020] [Accepted: 12/04/2020] [Indexed: 01/29/2023]
Abstract
BACKGROUND The most common thoracic injury in children, resulting in trauma, is pulmonary contusion (PC). Bone marrow-derived mesenchymal stem cells (BM-MSCs) are used in wound healing and many other diseases. This study aims to examine the effects of BM-MSCs on PC healing in rats. MATERIALS AND METHODS A total of 45 male Wistar albino rats were used. Four groups were formed. BM-MSCs were labeled with the green fluorescent protein. PC was observed in the control group. In group II, PC occured and left to spontaneous healing. In group III, PC formed and BM-MSCs were given. In group IV, BM-MSCs were given without PC formation. Subjects were sacrificed 1 week later. Whether there was any difference in terms of BM-MSC involvement and lung injury score was investigated. Statistical analysis was performed using the Statistical Package for Social Sciences (SPSS), version 17.0, software (SPSS Inc., Chicago, IL), and p value of <0.05 was considered statistically significant. RESULTS BM-MSCs were collected much more in the lungs in group III than in group IV. Group III had a lower lung injury score value than group II. CONCLUSION The greater involvement of the BM-MSCs in the injury site, and further reductions in lung injury score suggest that BM-MSCs are contributing to the healing of the injury. The use of BM-MSCs in risky patients with diffuse PC may be an alternative treatment to conventional methods.
Collapse
Affiliation(s)
- Sabri Demir
- Department of Pediatric Surgery, Ankara Bilkent City Hospital, Children Hospital, Ankara, Turkey; Department of Pediatric Surgery, School of Medicine, Kirikkale University, Kirikkale, Turkey.
| | - Ahmet Erturk
- Department of Pediatric Surgery, Ankara Bilkent City Hospital, Children Hospital, Ankara, Turkey; Department of Pediatric Surgery, School of Medicine, Kirikkale University, Kirikkale, Turkey
| | - Yasemin Dere Gunal
- Department of Pediatric Surgery, School of Medicine, Kirikkale University, Kirikkale, Turkey
| | - Ismail Ozmen
- Department of Pediatric Surgery, School of Medicine, Kirikkale University, Kirikkale, Turkey
| | - Mehmet Zengin
- Department of Pathology, School of Medicine, Kirikkale University, Kirikkale, Turkey
| | - Dincer Yildiz
- Faculty of Veterinary, Department of Anatomy, Kirikkale University, Kirikkale, Turkey
| | - Erdal Karaoz
- Department of Histology and Embryology, School of Medicine, Istinye University, İstanbul, Turkey
| | - Siyami Karahan
- Faculty of Veterinary, Department of Histology, Kirikkale University, Kirikkale, Turkey
| | - Emrah Senel
- Department of Pediatric Surgery, School of Medicine, Ankara Yıldırım Beyazıt University, Ankara, Turkey
| |
Collapse
|
16
|
Paul S, Candelario-Jalil E. Emerging neuroprotective strategies for the treatment of ischemic stroke: An overview of clinical and preclinical studies. Exp Neurol 2020; 335:113518. [PMID: 33144066 DOI: 10.1016/j.expneurol.2020.113518] [Citation(s) in RCA: 331] [Impact Index Per Article: 82.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/20/2020] [Accepted: 10/23/2020] [Indexed: 12/12/2022]
Abstract
Stroke is the leading cause of disability and thesecond leading cause of death worldwide. With the global population aged 65 and over growing faster than all other age groups, the incidence of stroke is also increasing. In addition, there is a shift in the overall stroke burden towards younger age groups, particularly in low and middle-income countries. Stroke in most cases is caused due to an abrupt blockage of an artery (ischemic stroke), but in some instances stroke may be caused due to bleeding into brain tissue when a blood vessel ruptures (hemorrhagic stroke). Although treatment options for stroke are still limited, with the advancement in recanalization therapy using both pharmacological and mechanical thrombolysis some progress has been made in helping patients recover from ischemic stroke. However, there is still a substantial need for the development of therapeutic agents for neuroprotection in acute ischemic stroke to protect the brain from damage prior to and during recanalization, extend the therapeutic time window for intervention and further improve functional outcome. The current review has assessed the past challenges in developing neuroprotective strategies, evaluated the recent advances in clinical trials, discussed the recent initiative by the National Institute of Neurological Disorders and Stroke in USA for the search of novel neuroprotectants (Stroke Preclinical Assessment Network, SPAN) and identified emerging neuroprotectants being currently evaluated in preclinical studies. The underlying molecular mechanism of each of the neuroprotective strategies have also been summarized, which could assist in the development of future strategies for combinational therapy in stroke treatment.
Collapse
Affiliation(s)
- Surojit Paul
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| | - Eduardo Candelario-Jalil
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
17
|
Gussenhoven R, Ophelders DRMG, Dudink J, Pieterman K, Lammens M, Mays RW, Zimmermann LJ, Kramer BW, Wolfs TGAM, Jellema RK. Systemic multipotent adult progenitor cells protect the cerebellum after asphyxia in fetal sheep. Stem Cells Transl Med 2020; 10:57-67. [PMID: 32985793 PMCID: PMC7780812 DOI: 10.1002/sctm.19-0157] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 06/29/2020] [Accepted: 08/09/2020] [Indexed: 12/30/2022] Open
Abstract
Involvement of the cerebellum in the pathophysiology of hypoxic‐ischemic encephalopathy (HIE) in preterm infants is increasingly recognized. We aimed to assess the neuroprotective potential of intravenously administered multipotent adult progenitor cells (MAPCs) in the preterm cerebellum. Instrumented preterm ovine fetuses were subjected to transient global hypoxia‐ischemia (HI) by 25 minutes of umbilical cord occlusion at 0.7 of gestation. After reperfusion, two doses of MAPCs were administered intravenously. MAPCs are a plastic adherent bone‐marrow‐derived population of adult progenitor cells with neuroprotective potency in experimental and clinical studies. Global HI caused marked cortical injury in the cerebellum, histologically indicated by disruption of cortical strata, impeded Purkinje cell development, and decreased dendritic arborization. Furthermore, global HI induced histopathological microgliosis, hypomyelination, and disruption of white matter organization. MAPC treatment significantly prevented cortical injury and region‐specifically attenuated white matter injury in the cerebellum following global HI. Diffusion tensor imaging (DTI) detected HI‐induced injury and MAPC neuroprotection in the preterm cerebellum. This study has demonstrated in a preclinical large animal model that early systemic MAPC therapy improved structural injury of the preterm cerebellum following global HI. Microstructural improvement was detectable with DTI. These findings support the potential of MAPC therapy for the treatment of HIE and the added clinical value of DTI for the detection of cerebellar injury and the evaluation of cell‐based therapy.
Collapse
Affiliation(s)
- Ruth Gussenhoven
- Department of Pediatrics, Maastricht University Medical Centre, Maastricht, The Netherlands.,School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Daan R M G Ophelders
- Department of Pediatrics, Maastricht University Medical Centre, Maastricht, The Netherlands.,School of Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Jeroen Dudink
- Department of Neonatology, Wilhelmina Children's Hospital and Brain Centre Rudolf Magnus, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Kay Pieterman
- Biomedical Imaging Group Rotterdam, Department of Radiology and Medical Informatics, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Martin Lammens
- Department of Pathology, Antwerp University Hospital and University of Antwerp, Edegem, Belgium
| | - Robert W Mays
- Regenerative Medicine, Athersys, Inc., Cleveland, Ohio, USA
| | - Luc J Zimmermann
- Department of Pediatrics, Maastricht University Medical Centre, Maastricht, The Netherlands.,School of Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Boris W Kramer
- Department of Pediatrics, Maastricht University Medical Centre, Maastricht, The Netherlands.,School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands.,School of Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Tim G A M Wolfs
- Department of Pediatrics, Maastricht University Medical Centre, Maastricht, The Netherlands.,School of Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Reint K Jellema
- Department of Pediatrics, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
18
|
Ophelders DR, Gussenhoven R, Klein L, Jellema RK, Westerlaken RJ, Hütten MC, Vermeulen J, Wassink G, Gunn AJ, Wolfs TG. Preterm Brain Injury, Antenatal Triggers, and Therapeutics: Timing Is Key. Cells 2020; 9:E1871. [PMID: 32785181 PMCID: PMC7464163 DOI: 10.3390/cells9081871] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/31/2020] [Accepted: 08/05/2020] [Indexed: 02/08/2023] Open
Abstract
With a worldwide incidence of 15 million cases, preterm birth is a major contributor to neonatal mortality and morbidity, and concomitant social and economic burden Preterm infants are predisposed to life-long neurological disorders due to the immaturity of the brain. The risks are inversely proportional to maturity at birth. In the majority of extremely preterm infants (<28 weeks' gestation), perinatal brain injury is associated with exposure to multiple inflammatory perinatal triggers that include antenatal infection (i.e., chorioamnionitis), hypoxia-ischemia, and various postnatal injurious triggers (i.e., oxidative stress, sepsis, mechanical ventilation, hemodynamic instability). These perinatal insults cause a self-perpetuating cascade of peripheral and cerebral inflammation that plays a critical role in the etiology of diffuse white and grey matter injuries that underlies a spectrum of connectivity deficits in survivors from extremely preterm birth. This review focuses on chorioamnionitis and hypoxia-ischemia, which are two important antenatal risk factors for preterm brain injury, and highlights the latest insights on its pathophysiology, potential treatment, and future perspectives to narrow the translational gap between preclinical research and clinical applications.
Collapse
Affiliation(s)
- Daan R.M.G. Ophelders
- Department of Pediatrics, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands; (D.R.M.G.O.); (R.G.); (L.K.); (R.K.J.); (R.J.J.W.); (M.C.H.)
- School for Oncology and Developmental Biology (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Ruth Gussenhoven
- Department of Pediatrics, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands; (D.R.M.G.O.); (R.G.); (L.K.); (R.K.J.); (R.J.J.W.); (M.C.H.)
| | - Luise Klein
- Department of Pediatrics, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands; (D.R.M.G.O.); (R.G.); (L.K.); (R.K.J.); (R.J.J.W.); (M.C.H.)
- School for Mental Health and Neuroscience (MHeNS), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Reint K. Jellema
- Department of Pediatrics, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands; (D.R.M.G.O.); (R.G.); (L.K.); (R.K.J.); (R.J.J.W.); (M.C.H.)
| | - Rob J.J. Westerlaken
- Department of Pediatrics, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands; (D.R.M.G.O.); (R.G.); (L.K.); (R.K.J.); (R.J.J.W.); (M.C.H.)
- School for Oncology and Developmental Biology (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Matthias C. Hütten
- Department of Pediatrics, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands; (D.R.M.G.O.); (R.G.); (L.K.); (R.K.J.); (R.J.J.W.); (M.C.H.)
- School for Oncology and Developmental Biology (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Jeroen Vermeulen
- Department of Pediatric Neurology, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands;
| | - Guido Wassink
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Private bag 92019, Auckland 1023, New Zealand; (G.W.); (A.J.G.)
| | - Alistair J. Gunn
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Private bag 92019, Auckland 1023, New Zealand; (G.W.); (A.J.G.)
| | - Tim G.A.M. Wolfs
- Department of Pediatrics, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands; (D.R.M.G.O.); (R.G.); (L.K.); (R.K.J.); (R.J.J.W.); (M.C.H.)
- School for Oncology and Developmental Biology (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
19
|
Atkinson SP. A preview of selected articles. Stem Cells Transl Med 2020. [PMCID: PMC7381805 DOI: 10.1002/sctm.20-0277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
20
|
Caplan HW, Prabhakara KS, Kumar A, Toledano‐Furman NE, Martin C, Carrillo L, Moreno NF, Bordt AS, Olson SD, Cox CS. Human cord blood-derived regulatory T-cell therapy modulates the central and peripheral immune response after traumatic brain injury. Stem Cells Transl Med 2020; 9:903-916. [PMID: 32383348 PMCID: PMC7381810 DOI: 10.1002/sctm.19-0444] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 03/13/2020] [Accepted: 04/04/2020] [Indexed: 12/19/2022] Open
Abstract
Traumatic brain injury (TBI) causes a profound inflammatory response within the central nervous system and peripheral immune system, which contributes to secondary brain injury and further morbidity and mortality. Preclinical investigations have demonstrated that treatments that downregulate microglia activation and polarize them toward a reparative/anti-inflammatory phenotype have improved outcomes in preclinical models. However, no therapy to date has translated into proven benefits in human patients. Regulatory T cells (Treg) have been shown to downregulate pathologic immune responses of the innate and adaptive immune system across a variety of pathologies. Furthermore, cellular therapy has been shown to augment host Treg responses in preclinical models; yet, studies investigating the use of Treg as a therapeutic for TBI are lacking. In a rodent TBI model, we demonstrate that human umbilical cord blood Treg modulate the central and peripheral immune response after injury in vitro and in vivo.
Collapse
Affiliation(s)
- Henry W. Caplan
- Department of Pediatric Surgery, McGovern Medical SchoolUniversity of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Karthik S. Prabhakara
- Department of Pediatric Surgery, McGovern Medical SchoolUniversity of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Akshita Kumar
- Department of Pediatric Surgery, McGovern Medical SchoolUniversity of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Naama E. Toledano‐Furman
- Department of Pediatric Surgery, McGovern Medical SchoolUniversity of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Cecilia Martin
- Department of Pediatric Surgery, McGovern Medical SchoolUniversity of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Louis Carrillo
- Department of Pediatric Surgery, McGovern Medical SchoolUniversity of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Nicolas F. Moreno
- Department of Pediatric Surgery, McGovern Medical SchoolUniversity of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Andrea S. Bordt
- Department of Pediatric Surgery, McGovern Medical SchoolUniversity of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Scott D. Olson
- Department of Pediatric Surgery, McGovern Medical SchoolUniversity of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Charles S. Cox
- Department of Pediatric Surgery, McGovern Medical SchoolUniversity of Texas Health Science Center at HoustonHoustonTexasUSA
| |
Collapse
|
21
|
Diaz MF, Horton PD, Kumar A, Livingston M, Mohammadalipour A, Xue H, Skibber MA, Ewere A, Toledano Furman NE, Aroom KR, Zhang S, Gill BS, Cox CS, Wenzel PL. Injury intensifies T cell mediated graft-versus-host disease in a humanized model of traumatic brain injury. Sci Rep 2020; 10:10729. [PMID: 32612177 PMCID: PMC7330041 DOI: 10.1038/s41598-020-67723-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 06/10/2020] [Indexed: 12/29/2022] Open
Abstract
The immune system plays critical roles in promoting tissue repair during recovery from neurotrauma but is also responsible for unchecked inflammation that causes neuronal cell death, systemic stress, and lethal immunodepression. Understanding the immune response to neurotrauma is an urgent priority, yet current models of traumatic brain injury (TBI) inadequately recapitulate the human immune response. Here, we report the first description of a humanized model of TBI and show that TBI places significant stress on the bone marrow. Hematopoietic cells of the marrow are regionally decimated, with evidence pointing to exacerbation of underlying graft-versus-host disease (GVHD) linked to presence of human T cells in the marrow. Despite complexities of the humanized mouse, marrow aplasia caused by TBI could be alleviated by cell therapy with human bone marrow mesenchymal stromal cells (MSCs). We conclude that MSCs could be used to ameliorate syndromes triggered by hypercytokinemia in settings of secondary inflammatory stimulus that upset marrow homeostasis such as TBI. More broadly, this study highlights the importance of understanding how underlying immune disorders including immunodepression, autoimmunity, and GVHD might be intensified by injury.
Collapse
Affiliation(s)
- Miguel F Diaz
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.,Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.,Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Paulina D Horton
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.,Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.,Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Akshita Kumar
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Megan Livingston
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.,Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.,Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Amina Mohammadalipour
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Hasen Xue
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Max A Skibber
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.,Department of Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Adesuwa Ewere
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.,Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.,School of Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Naama E Toledano Furman
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Kevin R Aroom
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Songlin Zhang
- Department of Pathology and Laboratory Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Brijesh S Gill
- Department of Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Charles S Cox
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.,Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Pamela L Wenzel
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA. .,Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA. .,Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| |
Collapse
|
22
|
Laing RW, Stubblefield S, Wallace L, Roobrouck VD, Bhogal RH, Schlegel A, Boteon YL, Reynolds GM, Ting AE, Mirza DF, Newsome PN, Mergental H, Afford SC. The Delivery of Multipotent Adult Progenitor Cells to Extended Criteria Human Donor Livers Using Normothermic Machine Perfusion. Front Immunol 2020; 11:1226. [PMID: 32714318 PMCID: PMC7344318 DOI: 10.3389/fimmu.2020.01226] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/15/2020] [Indexed: 12/30/2022] Open
Abstract
Background: Pre-clinical research with multi-potent adult progenitor cells (MAPC® cells, Multistem, Athersys Inc., Cleveland, Ohio) suggests their potential as an anti-inflammatory and immunomodulatory therapy in organ transplantation. Normothermic machine perfusion of the liver (NMP-L) has been proposed as a way of introducing therapeutic agents into the donor organ. Delivery of cellular therapy to human donor livers using this technique has not yet been described in the literature. The primary objectives of this study were to develop a technique for delivering cellular therapy to human donor livers using NMP-L and demonstrate engraftment. Methods: Six discarded human livers were perfused for 6 h at 37°C using the Liver Assist (Organ Assist, Groningen). 50 × 106 CMPTX-labeled MAPC cells were infused directly into the right lobe via the hepatic artery (HA, n = 3) or portal vein (PV, n = 3) over 20 min at different time points during the perfusion. Perfusion parameters were recorded and central and peripheral biopsies were taken at multiple time-points from both lobes and subjected to standard histological stains and confocal microscopy. Perfusate was analyzed using a 35-plex multiplex assay and proteomic analysis. Results: There was no detrimental effect on perfusion flow parameters on infusion of MAPC cells by either route. Three out of six livers met established criteria for organ viability. Confocal microscopy demonstrated engraftment of MAPC cells across vascular endothelium when perfused via the artery. 35-plex multiplex analysis of perfusate yielded 13 positive targets, 9 of which appeared to be related to the infusion of MAPC cells (including Interleukin's 1b, 4, 5, 6, 8, 10, MCP-1, GM-CSF, SDF-1a). Proteomic analysis revealed 295 unique proteins in the perfusate from time-points following the infusion of cellular therapy, many of which have strong links to MAPC cells and mesenchymal stem cells in the literature. Functional enrichment analysis demonstrated their immunomodulatory potential. Conclusion: We have demonstrated that cells can be delivered directly to the target organ, prior to host immune cell population exposure and without compromising the perfusion. Transendothelial migration occurs following arterial infusion. MAPC cells appear to secrete a host of soluble factors that would have anti-inflammatory and immunomodulatory benefits in a human model of liver transplantation.
Collapse
Affiliation(s)
- Richard W Laing
- NIHR Liver Biomedical Research Unit, Centre for Liver Research, College of Medical and Dental Sciences, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom.,Liver Unit, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | | | - Lorraine Wallace
- NIHR Liver Biomedical Research Unit, Centre for Liver Research, College of Medical and Dental Sciences, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | | | - Ricky H Bhogal
- NIHR Liver Biomedical Research Unit, Centre for Liver Research, College of Medical and Dental Sciences, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom.,Liver Unit, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Andrea Schlegel
- Liver Unit, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Yuri L Boteon
- NIHR Liver Biomedical Research Unit, Centre for Liver Research, College of Medical and Dental Sciences, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom.,Liver Unit, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Gary M Reynolds
- NIHR Liver Biomedical Research Unit, Centre for Liver Research, College of Medical and Dental Sciences, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | | | - Darius F Mirza
- NIHR Liver Biomedical Research Unit, Centre for Liver Research, College of Medical and Dental Sciences, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom.,Liver Unit, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Philip N Newsome
- NIHR Liver Biomedical Research Unit, Centre for Liver Research, College of Medical and Dental Sciences, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom.,Liver Unit, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Hynek Mergental
- NIHR Liver Biomedical Research Unit, Centre for Liver Research, College of Medical and Dental Sciences, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom.,Liver Unit, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Simon C Afford
- NIHR Liver Biomedical Research Unit, Centre for Liver Research, College of Medical and Dental Sciences, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom.,Liver Unit, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| |
Collapse
|
23
|
Zhu MM, Lin JH, Qing P, Pu L, Chen SL, Lin SJ, Li CL, Cao LX, Zhang YM. Manual acupuncture relieves microglia-mediated neuroinflammation in a rat model of traumatic brain injury by inhibiting the RhoA/ROCK2 pathway. Acupunct Med 2020; 38:426-434. [DOI: 10.1177/0964528420912248] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective: To investigate the regulatory mechanism of manual acupuncture (MA) on microglial polarization–mediated neuroinflammation after traumatic brain injury (TBI), focusing on the RhoA/Rho-associated coiled coil-forming protein kinase (ROCK2) pathway. Methods: Sprague Dawley (SD) rats were used to generate a TBI model using Feeney’s freefall epidural impact method. MA was performed on half of the TBI model rats, while the others remained untreated. Acupuncture was administered at GV15, GV16, GV20, GV26, and LI4. At the end of the intervention, rat brain tissue samples were collected, and the microglial M1 polarization status was observed by immunofluorescence labeling of CD86, an M1 microglia-specific protein. RhoA/ROCK2 signaling components were detected by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting. An enzyme-linked immunosorbent assay (ELISA) was used to detect the expression levels of inflammatory factors. Results: Compared with normal rats, the CD86 expression density in the untreated TBI model rats was high and showed an aggregated expression pattern. The genes and proteins of the RhoA/ROCK2 signaling pathway were highly expressed, and inflammatory factors were significantly increased. The CD86 expression density in TBI rats after MA was reduced compared to that in untreated TBI rats and showed a scattered distribution. The expression of RhoA/ROCK2 signaling pathway genes and proteins was also significantly reduced, and inflammatory factors were decreased. Conclusion: These results show that MA may inhibit M1 polarization of microglia by regulating the RhoA/ROCK2 signaling pathway, thereby reducing neuroinflammation in TBI.
Collapse
Affiliation(s)
- Ming-min Zhu
- Traditional Chinese Medical College of Jinan University, Guangzhou, China
| | - Ji-huan Lin
- Traditional Chinese Medical College of Jinan University, Guangzhou, China
| | - Peng Qing
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Liu Pu
- Traditional Chinese Medical College of Jinan University, Guangzhou, China
| | - Shu-lian Chen
- Traditional Chinese Medical College of Jinan University, Guangzhou, China
| | - Shu-jun Lin
- Traditional Chinese Medical College of Jinan University, Guangzhou, China
| | - Cheng-lu Li
- Traditional Chinese Medical College of Jinan University, Guangzhou, China
| | - Lu-xi Cao
- Traditional Chinese Medical College of Jinan University, Guangzhou, China
| | - Yi-min Zhang
- Traditional Chinese Medical College of Jinan University, Guangzhou, China
| |
Collapse
|
24
|
Caplan HW, Cardenas F, Gudenkauf F, Zelnick P, Xue H, Cox CS, Bedi SS. Spatiotemporal Distribution of Microglia After Traumatic Brain Injury in Male Mice. ASN Neuro 2020; 12:1759091420911770. [PMID: 32146827 PMCID: PMC7066592 DOI: 10.1177/1759091420911770] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Traumatic brain injury (TBI) disrupts the complex arrangement of glia and neuronal cells in the central nervous system. Microglia, the resident immune cells, survey the cellular milieu under homeostatic conditions and play a neuroprotective role via clearance of dead cells and debris such as axons and myelin. Resting (ramified) microglia possess a distinct morphology—small rod-shaped somata with thin processes. After TBI, microglia are activated and transition into an amoeboid morphology. To delineate the spatiotemporal morphological response of microglia after TBI, we used a controlled cortical impact injury model to quantify and characterize microglia at 24 hr and 28 days after TBI in the hippocampus (H) and lateral posterior nucleus of the thalamus (LPNT). Increased numbers of microglia were observed in the H and LPNT at 28 days after controlled cortical impact, but not at 24 hr in comparison to controls. Spatially, controlled cortical impact resulted in an increase of amoeboid microglia bilaterally at 24 hr and 28 days in H and ipsilaterally in LPNT. Temporally, at 28 days, TBI resulted in a significant increase in the number of amoeboid microglia in both H and LPNT. In addition, at 28 days after injury, we observed an increase in translocator protein, a marker for activated microglia, in the ipsilateral thalamus only. TBI results in a spatiotemporal increase in amoeboid microglia in the hippocampus and the LPNT over 28 days. Delineating their spatiotemporal phenotype is critical because it can help identify therapeutic targets with appropriate therapy.
Collapse
Affiliation(s)
- Henry W Caplan
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston
| | - Fanni Cardenas
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston
| | - Franciska Gudenkauf
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston
| | - Pamela Zelnick
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston
| | - Hasen Xue
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston
| | - Charles S Cox
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston
| | - Supinder S Bedi
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston
| |
Collapse
|
25
|
Therajaran P, Hamilton JA, O'Brien TJ, Jones NC, Ali I. Microglial polarization in posttraumatic epilepsy: Potential mechanism and treatment opportunity. Epilepsia 2020; 61:203-215. [PMID: 31943156 DOI: 10.1111/epi.16424] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 12/17/2022]
Abstract
Owing to the complexity of the pathophysiological mechanisms driving epileptogenesis following traumatic brain injury (TBI), effective preventive treatment approaches are not yet available for posttraumatic epilepsy (PTE). Neuroinflammation appears to play a critical role in the pathogenesis of the acquired epilepsies, including PTE, but despite a large preclinical literature demonstrating the ability of anti-inflammatory treatments to suppress epileptogenesis and chronic seizures, no anti-inflammatory treatment approaches have been clinically proven to date. TBI triggers robust inflammatory cascades, suggesting that they may be relevant for the pathogenesis of PTE. A major cell type involved in such cascades is the microglial cells-brain-resident immune cells that become activated after brain injury. When activated, these cells can oscillate between different phenotypes, and such polarization states are associated with the release of various pro- and anti-inflammatory mediators that may influence brain repair processes, and also differentially contribute to the development of PTE. As the molecular mechanisms and key signaling molecules associated with microglial polarization in brain are discovered, strategies are now emerging that can modulate this polarization, promoting this as a potential therapeutic strategy for PTE. In this review, we discuss the relevant literature regarding the polarization of brain-resident immune cells following TBI and attempt to put into perspective a role in epilepsy pathogenesis. Finally, we explore potential strategies that could polarize microglia/macrophages toward a neuroprotective phenotype to mitigate PTE development.
Collapse
Affiliation(s)
- Peravina Therajaran
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia
| | - John A Hamilton
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia
| | - Terence J O'Brien
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia.,Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Neurology, Alfred Hospital, Melbourne, Victoria, Australia
| | - Nigel C Jones
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia.,Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Neurology, Alfred Hospital, Melbourne, Victoria, Australia
| | - Idrish Ali
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia.,Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Neurology, Alfred Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
26
|
Bugay V, Bozdemir E, Vigil FA, Chun SH, Holstein DM, Elliott WR, Sprague CJ, Cavazos JE, Zamora DO, Rule G, Shapiro MS, Lechleiter JD, Brenner R. A Mouse Model of Repetitive Blast Traumatic Brain Injury Reveals Post-Trauma Seizures and Increased Neuronal Excitability. J Neurotrauma 2019; 37:248-261. [PMID: 31025597 DOI: 10.1089/neu.2018.6333] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Repetitive blast traumatic brain injury (TBI) affects numerous soldiers on the battlefield. Mild TBI has been shown to have long-lasting effects with repeated injury. We have investigated effects on neuronal excitability after repetitive, mild TBI in a mouse model of blast-induced brain injury. We exposed mice to mild blast trauma of an average peak overpressure of 14.6 psi, repeated across three consecutive days. While a single exposure did not reveal trauma as indicated by the glial fibrillary acidic protein indicator, three repetitive blasts did show significant increases. As well, mice had an increased indicator of inflammation (Iba-1) and increased tau, tau phosphorylation, and altered cytokine levels in the spleen. Video-electroencephalographic monitoring 48 h after the final blast exposure demonstrated seizures in 50% (12/24) of the mice, most of which were non-convulsive seizures. Long-term monitoring revealed that spontaneous seizures developed in at least 46% (6/13) of the mice. Patch clamp recording of dentate gyrus hippocampus neurons 48 h post-blast TBI demonstrated a shortened latency to the first spike and hyperpolarization of action potential threshold. We also found that evoked excitatory postsynaptic current amplitudes were significantly increased. These findings indicate that mild, repetitive blast exposures cause increases in neuronal excitability and seizures and eventual epilepsy development in some animals. The non-convulsive nature of the seizures suggests that subclinical seizures may occur in individuals experiencing even mild blast events, if repeated.
Collapse
Affiliation(s)
- Vladislav Bugay
- Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| | - Eda Bozdemir
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, Texas
| | - Fabio A Vigil
- Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| | - Sang H Chun
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, Texas
| | - Deborah M Holstein
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, Texas
| | - William R Elliott
- Sensory Trauma, United States Army Institute of Surgical Research, Fort Sam Houston San Antonio, Texas
| | - Cassie J Sprague
- Sensory Trauma, United States Army Institute of Surgical Research, Fort Sam Houston San Antonio, Texas
| | - Jose E Cavazos
- Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas.,Department of Neurology, University of Texas Health San Antonio, San Antonio, Texas
| | - David O Zamora
- Sensory Trauma, United States Army Institute of Surgical Research, Fort Sam Houston San Antonio, Texas
| | | | - Mark S Shapiro
- Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| | - James D Lechleiter
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, Texas
| | - Robert Brenner
- Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| |
Collapse
|
27
|
Khan RS, Newsome PN. A Comparison of Phenotypic and Functional Properties of Mesenchymal Stromal Cells and Multipotent Adult Progenitor Cells. Front Immunol 2019; 10:1952. [PMID: 31555259 PMCID: PMC6724467 DOI: 10.3389/fimmu.2019.01952] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 08/02/2019] [Indexed: 12/15/2022] Open
Abstract
Both Multipotent Adult Progenitor Cells and Mesenchymal Stromal Cells are bone-marrow derived, non-haematopoietic adherent cells, that are well-known for having immunomodulatory and pro-angiogenic properties, whilst being relatively non-immunogenic. However, they are phenotypically and functionally distinct cell types, which has implications for their efficacy in different settings. In this review we compare the phenotypic and functional properties of these two cell types, to help in determining which would be the superior cell type for different applications.
Collapse
Affiliation(s)
- Reenam S Khan
- National Institute for Health Research (NIHR), Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, United Kingdom.,Centre for Liver Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom.,Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Philip N Newsome
- National Institute for Health Research (NIHR), Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, United Kingdom.,Centre for Liver Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom.,Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| |
Collapse
|
28
|
Zhou Y, Shao A, Xu W, Wu H, Deng Y. Advance of Stem Cell Treatment for Traumatic Brain Injury. Front Cell Neurosci 2019; 13:301. [PMID: 31456663 PMCID: PMC6700304 DOI: 10.3389/fncel.2019.00301] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 06/19/2019] [Indexed: 01/01/2023] Open
Abstract
Traumatic brain injury (TBI) is an important cause of human mortality and morbidity, which can induce serious neurological damage. At present, clinical treatments for neurological dysfunction after TBI include hyperbaric oxygen, brain stimulation and behavioral therapy, but the therapeutic effect is not satisfactory. Recent studies have found that exogenous stem cells can migrate to damaged brain tissue, then participate in the repair of damaged brain tissue by further differentiation to replace damaged cells, while releasing anti-inflammatory factors and growth factors, thereby significantly improving neurological function. This article will mainly review the effects, deficiencies and related mechanisms of different types of stem cells in TBI.
Collapse
Affiliation(s)
- Yunxiang Zhou
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Weilin Xu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Haijian Wu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yongchuan Deng
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
29
|
Ouyang Q, Li F, Xie Y, Han J, Zhang Z, Feng Z, Su D, Zou X, Cai Y, Zou Y, Tang Y, Jiang X. Meta-Analysis of the Safety and Efficacy of Stem Cell Therapies for Ischemic Stroke in Preclinical and Clinical Studies. Stem Cells Dev 2019; 28:497-514. [PMID: 30739594 DOI: 10.1089/scd.2018.0218] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Affiliation(s)
- Qian Ouyang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Haizhu District, Guangzhou, China
| | - Feng Li
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Haizhu District, Guangzhou, China
| | - Yu Xie
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Haizhu District, Guangzhou, China
| | - Jianbang Han
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Haizhu District, Guangzhou, China
| | - Zhongfei Zhang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Haizhu District, Guangzhou, China
| | - Zhiming Feng
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Haizhu District, Guangzhou, China
| | - Dazhuang Su
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Haizhu District, Guangzhou, China
| | - Xiaoxiong Zou
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Haizhu District, Guangzhou, China
| | - Yingqian Cai
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Haizhu District, Guangzhou, China
| | - Yuxi Zou
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Haizhu District, Guangzhou, China
| | - Yanping Tang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Haizhu District, Guangzhou, China
| | - Xiaodan Jiang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Haizhu District, Guangzhou, China
| |
Collapse
|
30
|
Barkhuizen M, van Mechelen R, Vermeer M, Chedraui P, Paes D, van den Hove DL, Vaes B, Mays RW, Steinbusch HW, Robertson NJ, Kramer BW, Gavilanes AW. Systemic multipotent adult progenitor cells improve long-term neurodevelopmental outcomes after preterm hypoxic-ischemic encephalopathy. Behav Brain Res 2019; 362:77-81. [DOI: 10.1016/j.bbr.2019.01.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 12/28/2018] [Accepted: 01/09/2019] [Indexed: 11/16/2022]
|
31
|
Wang Z, He D, Zeng YY, Zhu L, Yang C, Lu YJ, Huang JQ, Cheng XY, Huang XH, Tan XJ. The spleen may be an important target of stem cell therapy for stroke. J Neuroinflammation 2019; 16:20. [PMID: 30700305 PMCID: PMC6352449 DOI: 10.1186/s12974-019-1400-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 01/07/2019] [Indexed: 12/21/2022] Open
Abstract
Stroke is the most common cerebrovascular disease, the second leading cause of death behind heart disease and is a major cause of long-term disability worldwide. Currently, systemic immunomodulatory therapy based on intravenous cells is attracting attention. The immune response to acute stroke is a major factor in cerebral ischaemia (CI) pathobiology and outcomes. Over the past decade, the significant contribution of the spleen to ischaemic stroke has gained considerable attention in stroke research. The changes in the spleen after stroke are mainly reflected in morphology, immune cells and cytokines, and these changes are closely related to the stroke outcomes. Autonomic nervous system (ANS) activation, release of central nervous system (CNS) antigens and chemokine/chemokine receptor interactions have been documented to be essential for efficient brain-spleen cross-talk after stroke. In various experimental models, human umbilical cord blood cells (hUCBs), haematopoietic stem cells (HSCs), bone marrow stem cells (BMSCs), human amnion epithelial cells (hAECs), neural stem cells (NSCs) and multipotent adult progenitor cells (MAPCs) have been shown to reduce the neurological damage caused by stroke. The different effects of these cell types on the interleukin (IL)-10, interferon (IFN), and cholinergic anti-inflammatory pathways in the spleen after stroke may promote the development of new cell therapy targets and strategies. The spleen will become a potential target of various stem cell therapies for stroke represented by MAPC treatment.
Collapse
Affiliation(s)
- Zhe Wang
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China.,Institute of Reproductive and Stem Cell Research, School of Basic Medical Science, Central South University, Changsha, 410000, China
| | - Da He
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China
| | - Ya-Yue Zeng
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China
| | - Li Zhu
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China
| | - Chao Yang
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China
| | - Yong-Juan Lu
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China
| | - Jie-Qiong Huang
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China
| | - Xiao-Yan Cheng
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China
| | - Xiang-Hong Huang
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China
| | - Xiao-Jun Tan
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China.
| |
Collapse
|
32
|
Peruzzaro ST, Andrews MMM, Al-Gharaibeh A, Pupiec O, Resk M, Story D, Maiti P, Rossignol J, Dunbar GL. Transplantation of mesenchymal stem cells genetically engineered to overexpress interleukin-10 promotes alternative inflammatory response in rat model of traumatic brain injury. J Neuroinflammation 2019; 16:2. [PMID: 30611291 PMCID: PMC6320578 DOI: 10.1186/s12974-018-1383-2] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 11/28/2018] [Indexed: 12/13/2022] Open
Abstract
Background Traumatic brain injury (TBI) is a major cause for long-term disability, yet the treatments available that improve outcomes after TBI limited. Neuroinflammatory responses are key contributors to determining patient outcomes after TBI. Transplantation of mesenchymal stem cells (MSCs), which release trophic and pro-repair cytokines, represents an effective strategy to reduce inflammation after TBI. One such pro-repair cytokine is interleukin-10 (IL-10), which reduces pro-inflammatory markers and trigger alternative inflammatory markers, such as CD163. In this study, we tested the therapeutic effects of MSCs that were engineered to overexpress IL-10 when transplanted into rats following TBI in the medial frontal cortex. Methods Thirty-six hours following TBI, rats were transplanted with MSCs and then assessed for 3 weeks on a battery of behavioral tests that measured motor and cognitive abilities. Histological evaluation was then done to measure the activation of the inflammatory response. Additionally, immunomodulatory effects were evaluated by immunohistochemistry and Western blot analyses. Results A significant improvement in fine motor function was observed in rats that received transplants of MSCs engineered to overexpress IL-10 (MSCs + IL-10) or MSCs alone compared to TBI + vehicle-treated rats. Although tissue spared was unchanged, anti-inflammatory effects were revealed by a reduction in the number of glial fibrillary acidic protein cells and CD86 cells in both TBI + MSCs + IL-10 and TBI + MSC groups compared to TBI + vehicle rats. Microglial activation was significantly increased in the TBI + MSC group when compared to the sham + vehicle group. Western blot data suggested a reduction in tumor necrosis factor-alpha in the TBI + MSCs + IL-10 group compared to TBI + MSC group. Immunomodulatory effects were demonstrated by a shift from classical inflammation expression (CD86) to an alternative inflammation state (CD163) in both treatments with MSCs and MSCs + IL-10. Furthermore, co-labeling of both CD86 and CD163 was detected in the same cells, suggesting a temporal change in macrophage expression. Conclusions Overall, our findings suggest that transplantation of MSCs that were engineered to overexpress IL-10 can improve functional outcomes by providing a beneficial perilesion environment. This improvement may be explained by the shifting of macrophage expression to a more pro-repair state, thereby providing a possible new therapy for treating TBI.
Collapse
Affiliation(s)
- S T Peruzzaro
- Field Neurosciences Institute of Laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI, 48859, USA.,Program in Neuroscience, Central Michigan University, Mt. Pleasant, MI, 48859, USA
| | - M M M Andrews
- Field Neurosciences Institute of Laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI, 48859, USA.,Program in Neuroscience, Central Michigan University, Mt. Pleasant, MI, 48859, USA
| | - A Al-Gharaibeh
- Field Neurosciences Institute of Laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI, 48859, USA.,Program in Neuroscience, Central Michigan University, Mt. Pleasant, MI, 48859, USA
| | - O Pupiec
- Field Neurosciences Institute of Laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI, 48859, USA.,Program in Neuroscience, Central Michigan University, Mt. Pleasant, MI, 48859, USA
| | - M Resk
- Field Neurosciences Institute of Laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI, 48859, USA.,Program in Neuroscience, Central Michigan University, Mt. Pleasant, MI, 48859, USA
| | - D Story
- Field Neurosciences Institute of Laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI, 48859, USA.,Program in Neuroscience, Central Michigan University, Mt. Pleasant, MI, 48859, USA.,Department of Psychology, Central Michigan University, Mt. Pleasant, MI, 48859, USA
| | - P Maiti
- Field Neurosciences Institute of Laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI, 48859, USA.,Program in Neuroscience, Central Michigan University, Mt. Pleasant, MI, 48859, USA.,Department of Psychology, Central Michigan University, Mt. Pleasant, MI, 48859, USA.,Field Neurosciences Institute, St. Mary's of Michigan, Saginaw, MI, 48604, USA.,Department of Biology, Saginaw Valley State University, Saginaw, MI, 48610, USA.,Brain Research Laboratory, Saginaw Valley State University, Saginaw, MI, 48610, USA
| | - J Rossignol
- Field Neurosciences Institute of Laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI, 48859, USA. .,Program in Neuroscience, Central Michigan University, Mt. Pleasant, MI, 48859, USA. .,College of Medicine, Central Michigan University, Mt. Pleasant, MI, 48859, USA.
| | - G L Dunbar
- Field Neurosciences Institute of Laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI, 48859, USA. .,Program in Neuroscience, Central Michigan University, Mt. Pleasant, MI, 48859, USA. .,Department of Psychology, Central Michigan University, Mt. Pleasant, MI, 48859, USA. .,Field Neurosciences Institute, St. Mary's of Michigan, Saginaw, MI, 48604, USA. .,Brain Research Laboratory, Saginaw Valley State University, Saginaw, MI, 48610, USA.
| |
Collapse
|
33
|
Prabhakara KS, Kota DJ, Jones GH, Srivastava AK, Cox CS, Olson SD. Teriflunomide Modulates Vascular Permeability and Microglial Activation after Experimental Traumatic Brain Injury. Mol Ther 2018; 26:2152-2162. [PMID: 30037655 PMCID: PMC6127507 DOI: 10.1016/j.ymthe.2018.06.022] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 06/22/2018] [Accepted: 06/25/2018] [Indexed: 12/18/2022] Open
Abstract
Despite intensive research and clinical trials with numerous therapeutic treatments, traumatic brain injury (TBI) is a serious public health problem in the United States. There is no effective FDA-approved treatment to reduce morbidity and mortality associated with TBI. Inflammation plays a pivotal role in the pathogenesis of TBI. We looked to re-purpose existing drugs that reduce immune activation without broad immunosuppression. Teriflunomide, an FDA-approved drug, has been shown to modulate immunological responses outside of its ability to inhibit pyrimidine synthesis in rapidly proliferating cells. In this study, we tested the efficacy of teriflunomide to treat two different injury intensities in rat models of TBI. Our results show that teriflunomide restores blood-brain barrier integrity, decreases inflammation, and increases neurogenesis in the subgranular zone of the hippocampus. While we were unable to detect neurocognitive effects of treatment on memory and special learning abilities after treatment, a 2-week treatment following injury was sufficient to reduce neuroinflammation up to 120 days later.
Collapse
Affiliation(s)
- Karthik S Prabhakara
- Program in Children's Regenerative Medicine, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Daniel J Kota
- Emory Personalized Immunotherapy Core Labs, Emory University, School of Medicine, Atlanta, GA 30322, USA
| | - Gregory H Jones
- Program in Children's Regenerative Medicine, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Amit K Srivastava
- Program in Children's Regenerative Medicine, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Charles S Cox
- Program in Children's Regenerative Medicine, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Scott D Olson
- Program in Children's Regenerative Medicine, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
34
|
Patel NA, Moss LD, Lee JY, Tajiri N, Acosta S, Hudson C, Parag S, Cooper DR, Borlongan CV, Bickford PC. Long noncoding RNA MALAT1 in exosomes drives regenerative function and modulates inflammation-linked networks following traumatic brain injury. J Neuroinflammation 2018; 15:204. [PMID: 30001722 PMCID: PMC6044101 DOI: 10.1186/s12974-018-1240-3] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 06/26/2018] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Neuroinflammation is a common therapeutic target for traumatic brain injury (TBI) due to its contribution to delayed secondary cell death and has the potential to occur for years after the initial insult. Exosomes from adipose-derived stem cells (hASCs) containing the long noncoding RNA MALAT1 are a novel, cell-free regenerative approach to long-term recovery after traumatic brain injury (TBI) that have the potential to modulate inflammation at the genomic level. The long noncoding RNA MALAT1 has been shown to be an important component of the secretome of hASCs. METHODS We isolated exosomes from hASC containing or depleted of MALAT1. The hASC-derived exosomes were then administered intravenously to rats following a mild controlled cortical impact (CCI). We followed the rats with behavior, in vivo imaging, histology, and RNA sequencing (RNA Seq). RESULTS Using in vivo imaging, we show that exosomes migrate into the spleen within 1 h following administration and enter the brain several hours later following TBI. Significant recovery of function on motor behavior as well as a reduction in cortical brain injury was observed after TBI in rats treated with exosomes. Treatment with either exosomes depleted of MALAT1 or conditioned media depleted of exosomes showed limited regenerative effects, demonstrating the importance of MALAT1 in exosome-mediated recovery. Analysis of the brain and spleen transcriptome using RNA Seq showed MALAT1-dependent modulation of inflammation-related pathways, cell cycle, cell death, and regenerative molecular pathways. Importantly, our data demonstrates that MALAT1 regulates expression of other noncoding RNAs including snoRNAs. CONCLUSION We demonstrate that MALAT1 in hASC-derived exosomes modulates multiple therapeutic targets, including inflammation, and has tremendous therapeutic potential for treatment of TBI.
Collapse
Affiliation(s)
- Niketa A. Patel
- 0000 0001 0624 9286grid.281075.9James A Haley Veterans Hospital, Research Service, Tampa, FL USA ,0000 0001 2353 285Xgrid.170693.aDepartment of Molecular Medicine, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612 USA
| | - Lauren Daly Moss
- 0000 0001 2353 285Xgrid.170693.aDepartment of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL USA
| | - Jea-Young Lee
- 0000 0001 2353 285Xgrid.170693.aDepartment of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL USA
| | - Naoki Tajiri
- 0000 0001 2353 285Xgrid.170693.aDepartment of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL USA ,0000 0001 0728 1069grid.260433.0Present address: Department of Neurophysiology & Brain Science, Graduate School of Medical Sciences & Medical School, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601 Japan
| | - Sandra Acosta
- 0000 0001 2353 285Xgrid.170693.aDepartment of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL USA
| | - Charles Hudson
- 0000 0001 0624 9286grid.281075.9James A Haley Veterans Hospital, Research Service, Tampa, FL USA
| | - Sajan Parag
- 0000 0001 2353 285Xgrid.170693.aDepartment of Molecular Medicine, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612 USA
| | - Denise R. Cooper
- 0000 0001 0624 9286grid.281075.9James A Haley Veterans Hospital, Research Service, Tampa, FL USA ,0000 0001 2353 285Xgrid.170693.aDepartment of Molecular Medicine, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612 USA
| | - Cesario V. Borlongan
- 0000 0001 2353 285Xgrid.170693.aDepartment of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL USA ,0000 0001 2353 285Xgrid.170693.aUSF Health Center of Excellence for Aging and Brain Repair MDC-78, 12901 Bruce B Downs, Blvd, Tampa, FL 33612 USA
| | - Paula C. Bickford
- 0000 0001 0624 9286grid.281075.9James A Haley Veterans Hospital, Research Service, Tampa, FL USA ,0000 0001 2353 285Xgrid.170693.aDepartment of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL USA ,0000 0001 2353 285Xgrid.170693.aUSF Health Center of Excellence for Aging and Brain Repair MDC-78, 12901 Bruce B Downs, Blvd, Tampa, FL 33612 USA
| |
Collapse
|
35
|
Mays RW, Savitz SI. Intravenous Cellular Therapies for Acute Ischemic Stroke. Stroke 2018; 49:1058-1065. [DOI: 10.1161/strokeaha.118.018287] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 03/01/2018] [Accepted: 03/08/2018] [Indexed: 02/07/2023]
Affiliation(s)
- Robert W. Mays
- From the Department of Neurosciences, Athersys, Inc, (R.W.M.)
| | - Sean I. Savitz
- Institute for Stroke and Cerebrovascular Disease, UTHealth, Houston, TX (S.I.S.)
| |
Collapse
|
36
|
Bedi SS, Aertker BM, Liao GP, Caplan HW, Bhattarai D, Mandy F, Mandy F, Fernandez LG, Zelnick P, Mitchell MB, Schiffer W, Johnson M, Denson E, Prabhakara K, Xue H, Smith P, Uray K, Olson SD, Mays RW, Cox CS. Therapeutic time window of multipotent adult progenitor therapy after traumatic brain injury. J Neuroinflammation 2018; 15:84. [PMID: 29548333 PMCID: PMC5856201 DOI: 10.1186/s12974-018-1122-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Accepted: 03/08/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) is a major cause of death and disability. TBI results in a prolonged secondary central neuro-inflammatory response. Previously, we have demonstrated that multiple doses (2 and 24 h after TBI) of multipotent adult progenitor cells (MAPC) delivered intravenously preserve the blood-brain barrier (BBB), improve spatial learning, and decrease activated microglia/macrophages in the dentate gyrus of the hippocampus. In order to determine if there is an optimum treatment window to preserve the BBB, improve cognitive behavior, and attenuate the activated microglia/macrophages, we administered MAPC at various clinically relevant intervals. METHODS We administered two injections intravenously of MAPC treatment at hours 2 and 24 (2/24), 6 and 24 (6/24), 12 and 36 (12/36), or 36 and 72 (36/72) post cortical contusion injury (CCI) at a concentration of 10 million/kg. For BBB experiments, animals that received MAPC at 2/24, 6/24, and 12/36 were euthanized 72 h post injury. The 36/72 treated group was harvested at 96 h post injury. RESULTS Administration of MAPC resulted in a significant decrease in BBB permeability when administered at 2/24 h after TBI only. For behavior experiments, animals were harvested post behavior paradigm. There was a significant improvement in spatial learning (120 days post injury) when compared to cortical contusion injury (CCI) in groups when MAPC was administered at or before 24 h. In addition, there was a significant decrease in activated microglia/macrophages in the dentate gyrus of hippocampus of the treated group (2/24) only when compared to CCI. CONCLUSIONS Intravenous injections of MAPC at or before 24 h after CCI resulted in improvement of the BBB, improved cognitive behavior, and attenuated activated microglia/macrophages in the dentate gyrus.
Collapse
Affiliation(s)
- Supinder S Bedi
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA.
| | - Benjamin M Aertker
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | - George P Liao
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | - Henry W Caplan
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | - Deepa Bhattarai
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | - Fanni Mandy
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | - Franciska Mandy
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | - Luis G Fernandez
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | - Pamela Zelnick
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | - Matthew B Mitchell
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | - Walter Schiffer
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | - Margaret Johnson
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | - Emma Denson
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | - Karthik Prabhakara
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | - Hasen Xue
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | - Philippa Smith
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | - Karen Uray
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | - Scott D Olson
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | | | - Charles S Cox
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA.,Departments of Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA.,Michael E DeBakey Institute for Comparative Cardiovascular Science and Biomedical Devices and Athersys, Inc., Cleveland, OH, USA
| |
Collapse
|
37
|
Gao J, Grill RJ, Dunn TJ, Bedi S, Labastida JA, Hetz RA, Xue H, Thonhoff JR, DeWitt DS, Prough DS, Cox CS, Wu P. Human Neural Stem Cell Transplantation-Mediated Alteration of Microglial/Macrophage Phenotypes after Traumatic Brain Injury. Cell Transplant 2018; 25:1863-1877. [PMID: 26980267 DOI: 10.3727/096368916x691150] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Neural stem cells (NSCs) promote recovery from brain trauma, but neuronal replacement is unlikely the sole underlying mechanism. We hypothesize that grafted NSCs enhance neural repair at least partially through modulating the host immune response after traumatic brain injury (TBI). C57BL/6 mice were intracerebrally injected with primed human NSCs (hNSCs) or vehicle 24 h after a severe controlled cortical impact injury. Six days after transplantation, brain tissues were collected for Western blot and immunohistochemical analyses. Observations included indicators of microglia/macrophage activation, M1 and M2 phenotypes, axonal injury detected by amyloid precursor protein (APP), lesion size, and the fate of grafted hNSCs. Animals receiving hNSC transplantation did not show significant decreases of brain lesion volumes compared to transplantation procedures with vehicle alone, but did show significantly reduced injury-dependent accumulation of APP. Furthermore, intracerebral transplantation of hNSCs reduced microglial activation as shown by a diminished intensity of Iba1 immunostaining and a transition of microglia/macrophages toward the M2 anti-inflammatory phenotype. The latter was represented by an increase in the brain M2/M1 ratio and increases of M2 microglial proteins. These phenotypic switches were accompanied by the increased expression of anti-inflammatory interleukin-4 receptor α and decreased proinflammatory interferon-γ receptor β. Finally, grafted hNSCs mainly differentiated into neurons and were phagocytized by either M1 or M2 microglia/macrophages. Thus, intracerebral transplantation of primed hNSCs efficiently leads host microglia/macrophages toward an anti-inflammatory phenotype that presumably contributes to stem cell-mediated neuroprotective effects after severe TBI in mice.
Collapse
Affiliation(s)
- Junling Gao
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Raymond J Grill
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Tiffany J Dunn
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Supinder Bedi
- Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, TX, USA
| | - Javier Allende Labastida
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Robert A Hetz
- Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, TX, USA
| | - Hasen Xue
- Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, TX, USA
| | - Jason R Thonhoff
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Douglas S DeWitt
- Department of Anesthesiology, University of Texas Medical Branch at Galveston, TX, USA
| | - Donald S Prough
- Department of Anesthesiology, University of Texas Medical Branch at Galveston, TX, USA
| | - Charles S Cox
- Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, TX, USA
| | - Ping Wu
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch at Galveston, Galveston, TX, USA.,Beijing Institute for Brain Disorders, Beijing, P.R. China
| |
Collapse
|
38
|
Abstract
Neurological injury is the primary lethal mechanism of injury in children, and the primary etiology of long-term disability after trauma. Laboratories and clinical/translational teams have sought to develop stem/progenitor cell therapies to improve recovery in a clinical setting in which there is no significant reparative option. While none of these treatments are currently standard therapeutics, phase IIb clinical trials are underway in both adults and children in severe traumatic brain injury (TBI) and phase I/IIa trials in spinal cord injury. This review will characterize the cell therapy strategies: cell replacement and tissue integration vs. immunomodulation/enhanced endogenous tissue repair. TBI is somewhat different from other central nervous system injuries (spinal cord injury and stroke), in that TBI is a diffuse injury, whereas spinal cord injury and stroke are anatomically discrete. Importantly, this drives cell therapy approaches, as TBI is less apt to be treatable with a local cell replacement intervention. More localized injuries may be more amenable to local approaches and cell replacement to bridge focal gaps. This review focuses on a few reports in the field that highlight areas of progress, but is not intended to be a comprehensive survey of the state of regenerative medicine for neurological injuries.
Collapse
|
39
|
Usage of Multiparameter Flow Cytometry to Study Microglia and Macrophage Heterogeneity in the Central Nervous System During Neuroinflammation and Neurodegeneration. Methods Mol Biol 2018; 1745:167-177. [PMID: 29476469 DOI: 10.1007/978-1-4939-7680-5_10] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The resident macrophages of the central nervous system (CNS), also known as microglia, and blood-derived macrophages play an important role in the functional activity of the normal CNS, as well as in the development of neuroinflammation during various neurodegenerative disorders. Microglia and macrophages represent heterogeneous populations, which can modulate CNS environment and have different effects on neuronal regeneration. In this chapter, the main features of microglial and macrophage subsets and current methods for investigation of their heterogeneity will be discussed.
Collapse
|
40
|
Guo H, Zhao N, Gao H, He X. Mesenchymal Stem Cells Overexpressing Interleukin-35 Propagate Immunosuppressive Effects in Mice. Scand J Immunol 2017; 86:389-395. [PMID: 28888053 DOI: 10.1111/sji.12613] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 09/04/2017] [Indexed: 12/20/2022]
Affiliation(s)
- H. Guo
- Department of General Surgery; Tianjin Medical University General Hospital; Tianjin China
- Tianjin General Surgery Institute; Tianjin Medical University General Hospital; Tianjin China
| | - N. Zhao
- Tianjin General Surgery Institute; Tianjin Medical University General Hospital; Tianjin China
| | - H. Gao
- Department of General Surgery; Tianjin Medical University General Hospital; Tianjin China
- Tianjin General Surgery Institute; Tianjin Medical University General Hospital; Tianjin China
| | - X. He
- Department of General Surgery; Tianjin Medical University General Hospital; Tianjin China
- Tianjin General Surgery Institute; Tianjin Medical University General Hospital; Tianjin China
| |
Collapse
|
41
|
Traumatic Brain Injury and Stem Cell: Pathophysiology and Update on Recent Treatment Modalities. Stem Cells Int 2017; 2017:6392592. [PMID: 28852409 PMCID: PMC5568618 DOI: 10.1155/2017/6392592] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 07/26/2017] [Indexed: 12/30/2022] Open
Abstract
Traumatic brain injury (TBI) is a complex condition that presents with a wide spectrum of clinical symptoms caused by an initial insult to the brain through an external mechanical force to the skull. In the United States alone, TBI accounts for more than 50,000 deaths per year and is one of the leading causes of mortality among young adults in the developed world. Pathophysiology of TBI is complex and consists of acute and delayed injury. In the acute phase, brain tissue destroyed upon impact includes neurons, glia, and endothelial cells, the latter of which makes up the blood-brain barrier. In the delayed phase, “toxins” released from damaged cells set off cascades in neighboring cells eventually leading to exacerbation of primary injury. As researches further explore pathophysiology and molecular mechanisms underlying this debilitating condition, numerous potential therapeutic strategies, especially those involving stem cells, are emerging to improve recovery and possibly reverse damage. In addition to elucidating the most recent advances in the understanding of TBI pathophysiology, this review explores two primary pathways currently under investigation and are thought to yield the most viable therapeutic approach for treatment of TBI: manipulation of endogenous neural cell response and administration of exogenous stem cell therapy.
Collapse
|
42
|
Crosstalk with Inflammatory Macrophages Shapes the Regulatory Properties of Multipotent Adult Progenitor Cells. Stem Cells Int 2017; 2017:2353240. [PMID: 28785285 PMCID: PMC5529661 DOI: 10.1155/2017/2353240] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 05/27/2017] [Accepted: 06/12/2017] [Indexed: 01/25/2023] Open
Abstract
Macrophages and microglia are key effector cells in immune-mediated neuroinflammatory disorders. Driving myeloid cells towards an anti-inflammatory, tissue repair-promoting phenotype is considered a promising strategy to halt neuroinflammation and promote central nervous system (CNS) repair. In this study, we defined the impact of multipotent adult progenitor cells (MAPC), a stem cell population sharing common mesodermal origin with mesenchymal stem cells (MSCs), on the phenotype of macrophages and the reciprocal interactions between these two cell types. We show that MAPC suppress the secretion of tumor necrosis factor alpha (TNF-α) by inflammatory macrophages partially through a cyclooxygenase 2- (COX-2-) dependent mechanism. In turn, we demonstrate that inflammatory macrophages trigger the immunomodulatory properties of MAPC, including an increased expression of immunomodulatory mediators (e.g., inducible nitric oxide synthase (iNOS) and COX-2), chemokines, and chemokine receptors. Macrophage-primed MAPC secrete soluble factors that suppress TNF-α release by macrophages. Moreover, the MAPC secretome suppresses the antigen-specific proliferation of autoreactive T cells and the T cell stimulatory capacity of macrophages. Finally, MAPC increase their motility towards secreted factors of activated macrophages. Collectively, these in vitro findings reveal intimate reciprocal interactions between MAPC and inflammatory macrophages, which are of importance in the design of MAPC-based therapeutic strategies for neuroinflammatory disorders in which myeloid cells play a crucial role.
Collapse
|
43
|
Abstract
Shibani Pati and Todd Rasmussen summarize progress in preclinical research on cellular therapeutics for traumatic injury and its sequelae and discuss prospects for clinical translation.
Collapse
|
44
|
Donat CK, Scott G, Gentleman SM, Sastre M. Microglial Activation in Traumatic Brain Injury. Front Aging Neurosci 2017; 9:208. [PMID: 28701948 PMCID: PMC5487478 DOI: 10.3389/fnagi.2017.00208] [Citation(s) in RCA: 282] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 06/12/2017] [Indexed: 12/15/2022] Open
Abstract
Microglia have a variety of functions in the brain, including synaptic pruning, CNS repair and mediating the immune response against peripheral infection. Microglia rapidly become activated in response to CNS damage. Depending on the nature of the stimulus, microglia can take a number of activation states, which correspond to altered microglia morphology, gene expression and function. It has been reported that early microglia activation following traumatic brain injury (TBI) may contribute to the restoration of homeostasis in the brain. On the other hand, if they remain chronically activated, such cells display a classically activated phenotype, releasing pro-inflammatory molecules, resulting in further tissue damage and contributing potentially to neurodegeneration. However, new evidence suggests that this classification is over-simplistic and the balance of activation states can vary at different points. In this article, we review the role of microglia in TBI, analyzing their distribution, morphology and functional phenotype over time in animal models and in humans. Animal studies have allowed genetic and pharmacological manipulations of microglia activation, in order to define their role. In addition, we describe investigations on the in vivo imaging of microglia using translocator protein (TSPO) PET and autoradiography, showing that microglial activation can occur in regions far remote from sites of focal injuries, in humans and animal models of TBI. Finally, we outline some novel potential therapeutic approaches that prime microglia/macrophages toward the beneficial restorative microglial phenotype after TBI.
Collapse
Affiliation(s)
| | | | | | - Magdalena Sastre
- Division of Brain Sciences, Department of Medicine, Imperial College LondonLondon, United Kingdom
| |
Collapse
|
45
|
Yang B, Hamilton JA, Valenzuela KS, Bogaerts A, Xi X, Aronowski J, Mays RW, Savitz SI. Multipotent Adult Progenitor Cells Enhance Recovery After Stroke by Modulating the Immune Response from the Spleen. Stem Cells 2017; 35:1290-1302. [PMID: 28263009 DOI: 10.1002/stem.2600] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 01/06/2017] [Accepted: 01/24/2017] [Indexed: 02/03/2023]
Abstract
Stem cell therapy modulates not only the local microenvironment of the brain but also the systemic immune responses. We explored the impact of human multipotent adult progenitor cells (MAPC) modulating splenic activation and peripheral immune responses after ischemic stroke. Hundred twenty-six Long-Evans adult male rats underwent middle cerebral artery occlusion. Twenty-four hours later, they received IV MAPC or saline treatment. At 3 days after infusion, RNA was isolated from the injured cortex and spleen for microarray analysis. Spleen mass, splenocyte phenotype, and releasing cytokines were measured. Serum cytokines, MAPC biodistribution, brain lesion sizes and neurofunctional deficits were compared in rats treated with MAPC or saline with and without spleens. Stroked animals treated with MAPC exhibited genes that more closely resembled animals with sham surgery. Gene categories downregulated by MAPC included leukocyte activation, antigen presentation, and immune effector processing, associated with the signaling pathways regulated by TNF-α, IL-1β, IL-6, and IFN-γ within the brain. MAPC treatment restored spleen mass reduction caused by stroke, elevated Treg cells within the spleen, increased IL-10 and decreased IL-1β released by splenocytes. MAPC reduced IL-6 and IL-1β and upregulated IL-10 serum levels. Compared with saline, MAPC enhance stroke recovery in rats with intact spleens but had no effects in rats without spleens. MAPC restores expression of multiple genes and pathways involved in immune and inflammatory responses after stroke. Immunomodulation of the splenic response by the intravenous administration of MAPC may create a more favorable environment for brain repair after stroke. Stem Cells 2017;35:1290-1302.
Collapse
Affiliation(s)
- Bing Yang
- Stroke Program, McGovern Medical School at UT-Health Houston, Houston, Texas, USA
| | - Jason A Hamilton
- Athersys, Cleveland, Ohio, USA.,Novartis Pharmaceutical Corp, East Hanover, New Jersey, USA
| | - Krystal S Valenzuela
- Stroke Program, McGovern Medical School at UT-Health Houston, Houston, Texas, USA
| | | | - XiaoPei Xi
- Stroke Program, McGovern Medical School at UT-Health Houston, Houston, Texas, USA
| | - Jaroslaw Aronowski
- Stroke Program, McGovern Medical School at UT-Health Houston, Houston, Texas, USA
| | | | - Sean I Savitz
- Stroke Program, McGovern Medical School at UT-Health Houston, Houston, Texas, USA
| |
Collapse
|
46
|
Kirby GTS, Mills SJ, Vandenpoel L, Pinxteren J, Ting A, Short RD, Cowin AJ, Michelmore A, Smith LE. Development of Advanced Dressings for the Delivery of Progenitor Cells. ACS APPLIED MATERIALS & INTERFACES 2017; 9:3445-3454. [PMID: 28068055 DOI: 10.1021/acsami.6b14725] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Culture surfaces that substantially reduce the degree of cell manipulation in the delivery of cell sheets to patients are described. These surfaces support the attachment, culture, and delivery of multipotent adult progenitor cells (MAPC). It was essential that the processes of attachment/detachment to the surface did not affect cell phenotype nor the function of the cultured cells. Both acid-based and amine-based surface coatings were generated from acrylic acid, propanoic acid, diaminopropane, and heptylamine precursors, respectively. While both functional groups supported cell attachment/detachment, amine coated surfaces gave optimal performance. X-ray photoelectron spectroscopy (XPS) showed that at a primary amine to carbon surface ratio of between 0.01 and 0.02, greater than 90% of attached cells were effectively transferred to a model wound bed. A dependence on primary amine concentration has not previously been reported. After 48 h of culture on the optimized amine surface, PCR, functional, and viability assays showed that MAPC retained their stem cell phenotype, full metabolic activity, and biological function. Consequently, in a proof of concept experiment, it was shown that this amine surface when coated onto a surgical dressing provides an effective and simple technology for the delivery of MAPC to murine dorsal excisional wounds, with MAPC delivery verified histologically. By optimizing for cell delivery using a combination of in vitro and in vivo techniques, we developed an effective surface for the delivery of MAPC in a clinically relevant format.
Collapse
Affiliation(s)
- Giles T S Kirby
- Cooperative Research Centre for Cell Therapy Manufacturing , North Terrace, Adelaide, South Australia 5000, Australia
- Future Industries Institute, University of South Australia , Mawson Lakes, South Australia 5095, Australia
| | - Stuart J Mills
- Cooperative Research Centre for Cell Therapy Manufacturing , North Terrace, Adelaide, South Australia 5000, Australia
- Future Industries Institute, University of South Australia , Mawson Lakes, South Australia 5095, Australia
| | - Liesbeth Vandenpoel
- Cooperative Research Centre for Cell Therapy Manufacturing , North Terrace, Adelaide, South Australia 5000, Australia
- ReGenesys BVBA, Bio-Incubator Leuven , Gaston Geenslaan 1, 3001 Heverlee, Belgium
| | - Jef Pinxteren
- Cooperative Research Centre for Cell Therapy Manufacturing , North Terrace, Adelaide, South Australia 5000, Australia
- ReGenesys BVBA, Bio-Incubator Leuven , Gaston Geenslaan 1, 3001 Heverlee, Belgium
| | - Anthony Ting
- Cooperative Research Centre for Cell Therapy Manufacturing , North Terrace, Adelaide, South Australia 5000, Australia
- Athersys, Inc. , Cleveland, Ohio 44115-2634, United States
| | - Robert D Short
- Cooperative Research Centre for Cell Therapy Manufacturing , North Terrace, Adelaide, South Australia 5000, Australia
- Future Industries Institute, University of South Australia , Mawson Lakes, South Australia 5095, Australia
| | - Allison J Cowin
- Cooperative Research Centre for Cell Therapy Manufacturing , North Terrace, Adelaide, South Australia 5000, Australia
- Future Industries Institute, University of South Australia , Mawson Lakes, South Australia 5095, Australia
| | - Andrew Michelmore
- Cooperative Research Centre for Cell Therapy Manufacturing , North Terrace, Adelaide, South Australia 5000, Australia
- Future Industries Institute, University of South Australia , Mawson Lakes, South Australia 5095, Australia
- School of Engineering, University of South Australia , Mawson Lakes, South Australia 5095, Australia
| | - Louise E Smith
- Cooperative Research Centre for Cell Therapy Manufacturing , North Terrace, Adelaide, South Australia 5000, Australia
- Future Industries Institute, University of South Australia , Mawson Lakes, South Australia 5095, Australia
| |
Collapse
|
47
|
The Polarization States of Microglia in TBI: A New Paradigm for Pharmacological Intervention. Neural Plast 2017; 2017:5405104. [PMID: 28255460 PMCID: PMC5309408 DOI: 10.1155/2017/5405104] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 12/17/2016] [Accepted: 01/11/2017] [Indexed: 01/04/2023] Open
Abstract
Traumatic brain injury (TBI) is a serious medical and social problem worldwide. Because of the complex pathophysiological mechanisms of TBI, effective pharmacotherapy is still lacking. The microglial cells are resident tissue macrophages located in the brain and have two major polarization states, M1 phenotype and M2 phenotype, when activated. The M1 phenotype is related to the release of proinflammatory cytokines and secondary brain injury, while the M2 phenotype has been proved to be responsible for the release of anti-inflammation cytokines and for central nervous system (CNS) repair. In animal models, pharmacological strategies inhibiting the M1 phenotype and promoting the M2 phenotype of microglial cells could alleviate cerebral damage and improve neurological function recovery after TBI. In this review, we aimed to summarize the current knowledge about the pathological significance of microglial M1/M2 polarization in the pathophysiology of TBI. In addition, we reviewed several drugs that have provided neuroprotective effects against brain injury following TBI by altering the polarization states of the microglia. We emphasized that future investigation of the regulation mechanisms of microglial M1/M2 polarization in TBI is anticipated, which could contribute to the development of new targets of pharmacological intervention in TBI.
Collapse
|
48
|
Matthay MA, Pati S, Lee JW. Concise Review: Mesenchymal Stem (Stromal) Cells: Biology and Preclinical Evidence for Therapeutic Potential for Organ Dysfunction Following Trauma or Sepsis. Stem Cells 2017; 35:316-324. [PMID: 27888550 DOI: 10.1002/stem.2551] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 11/07/2016] [Accepted: 11/08/2016] [Indexed: 12/12/2022]
Abstract
Several experimental studies have provided evidence that bone-marrow derived mesenchymal stem (stromal) cells (MSC) may be effective in treating critically ill surgical patients who develop traumatic brain injury, acute renal failure, or the acute respiratory distress syndrome. There is also preclinical evidence that MSC may be effective in treating sepsis-induced organ failure, including evidence that MSC have antimicrobial properties. This review considers preclinical studies with direct relevance to organ failure following trauma, sepsis or major infections that apply to critically ill patients. Progress has been made in understanding the mechanisms of benefit, including MSC release of paracrine factors, transfer of mitochondria, and elaboration of exosomes and microvesicles. Regardless of how well they are designed, preclinical studies have limitations in modeling the complexity of clinical syndromes, especially in patients who are critically ill. In order to facilitate translation of the preclinical studies of MSC to critically ill patients, there will need to be more standardization regarding MSC production with a focus on culture methods and cell characterization. Finally, well designed clinical trials will be needed in critically ill patient to assess safety and efficacy. Stem Cells 2017;35:316-324.
Collapse
Affiliation(s)
- Michael A Matthay
- Departments of Medicine and Anesthesia and the Cardiovascular Research Institute, University of California, San Francisco, USA
| | - Shibani Pati
- Department of Laboratory Medicine, University of California, Blood Systems Research Institute, San Francisco, USA
| | - Jae-Woo Lee
- Department of Anesthesia, University of California, San Francisco, USA
| |
Collapse
|
49
|
Miyauchi JT, Chen D, Choi M, Nissen JC, Shroyer KR, Djordevic S, Zachary IC, Selwood D, Tsirka SE. Ablation of Neuropilin 1 from glioma-associated microglia and macrophages slows tumor progression. Oncotarget 2016; 7:9801-14. [PMID: 26755653 PMCID: PMC4891085 DOI: 10.18632/oncotarget.6877] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 01/03/2016] [Indexed: 01/09/2023] Open
Abstract
Gliomas are the most commonly diagnosed primary tumors of the central nervous system (CNS). Median times of survival are dismal regardless of the treatment approach, underlying the need to develop more effective therapies. Modulation of the immune system is a promising strategy as innate and adaptive immunity play important roles in cancer progression. Glioma associated microglia and macrophages (GAMs) can comprise over 30% of the cells in glioma biopsies. Gliomas secrete cytokines that suppress the anti-tumorigenic properties of GAMs, causing them to secrete factors that support the tumor's spread and growth. Neuropilin 1 (Nrp1) is a transmembrane receptor that in mice both amplifies pro-angiogenic signaling in the tumor microenvironment and affects behavior of innate immune cells. Using a Cre-lox system, we generated mice that lack expression of Nrp1 in GAMs. We demonstrate, using an in vivo orthotopic glioma model, that tumors in mice with Nrp1-deficient GAMs exhibit less vascularity, grow at a slower pace, and are populated by increased numbers of anti-tumorigenic GAMs. Moreover, glioma survival times in mice with Nrp1-deficient GAMs were significantly longer. Treating wild-type mice with a small molecule inhibitor of Nrp1's b1 domain, EG00229, which we show here is selective for Nrp1 over Nrp2, yielded an identical outcome. Nrp1-deficient or EG00229-treated wild-type microglia exhibited a shift towards anti-tumorigenicity as evident by altered inflammatory marker profiles in vivo and decreased SMAD2/3 activation when conditioned in the presence of glioma-derived factors. These results provide support for the proposal that pharmacological inhibition of Nrp1 constitutes a potential strategy for suppressing glioma progression.
Collapse
Affiliation(s)
- Jeremy T Miyauchi
- Department of Pharmacology, Stony Brook University, Stony Brook, NY, USA
| | - Danling Chen
- Department of Pharmacology, Stony Brook University, Stony Brook, NY, USA
| | - Matthew Choi
- Department of Pharmacology, Stony Brook University, Stony Brook, NY, USA
| | - Jillian C Nissen
- Department of Pharmacology, Stony Brook University, Stony Brook, NY, USA
| | - Kenneth R Shroyer
- Department of Pathology, Stony Brook University, Stony Brook, NY, USA
| | - Snezana Djordevic
- Institute of Structural and Molecular Biology, University College London, London, UK
| | - Ian C Zachary
- Centre for Cardiovascular Biology and Medicine, Division of Medicine, University College London, London, UK
| | - David Selwood
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Stella E Tsirka
- Department of Pharmacology, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
50
|
Gao C, Qian Y, Huang J, Wang D, Su W, Wang P, Guo L, Quan W, An S, Zhang J, Jiang R. A Three-Day Consecutive Fingolimod Administration Improves Neurological Functions and Modulates Multiple Immune Responses of CCI Mice. Mol Neurobiol 2016; 54:8348-8360. [PMID: 27924525 DOI: 10.1007/s12035-016-0318-0] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 11/21/2016] [Indexed: 12/15/2022]
Abstract
Excessive inflammation after traumatic brain injury (TBI) is a major cause of secondary TBI. Though several inflammatory biomarkers have been postulated as the risk factors of TBI, there has not been any comprehensive description of them. Fingolimod, a new kind of immunomodulatory agent which can diminish various kinds of inflammatory responses, has shown additional therapeutic effects in the treatment of intracranial cerebral hematoma (ICH), ischemia, spinal cord injury (SCI), and many other CNS disorders. However, its therapeutic application has not been confirmed in TBI. Thus, we hypothesized that a 3-day consecutive fingolimod administration could broadly modulate the inflammatory reactions and improve the outcomes of TBI. The TBI models of C57/BL6 mice were established with the controlled cortical impact injury (CCI) system. A 3-day consecutive fingolimod therapy (given at 1, 24, and 48 h post injury) was performed at a dose of 1 mg/kg. The flow cytometry, immunoflourence, cytokine array, and ELISA were all applied to evaluate the immune cells and inflammatory markers in the injured brains. Immunohistochemical staining with anti-APP antibody was performed to assess the axonal damage. The neurological functions of these TBI models were assessed by mNSS/Rota-rod and Morris water maze (MWM). The brain water content and integrity of the blood-brain barrier (BBB) were also observed. On the 3rd day after TBI, the accumulation of inflammatory cytokines and chemokines reached the peak and administration of fingolimod reduced as many as 20 kinds of cytokines and chemokines. Fingolimod decreased infiltrated T lymphocytes and NK cells but increased the percentage of regulatory T (Treg) cells, and the concentration of IL-10 on the 3rd day after TBI. Fingolimod also notably attenuated the general activated microglia but augmented the M2/M1 ratio accompanied by decreased axonal damage. The neurological functions were improved after the fingolimod treatment accompanied with alleviation of the brain edema and BBB damage. This study suggests that the 3-day consecutive fingolimod administration extensively modulates multiple immuno-inflammatory responses and improves the neurological deficits after TBI, and therefore, it may be a new approach to the treatment of secondary TBI.
Collapse
Affiliation(s)
- Chuang Gao
- Department of Neurosurgery, Tianjin Medical University, General Hospital, Tianjin, China.,Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin, China
| | - Yu Qian
- Department of Neurosurgery, Tianjin Medical University, General Hospital, Tianjin, China.,Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin, China
| | - Jinhao Huang
- Department of Neurosurgery, Tianjin Medical University, General Hospital, Tianjin, China.,Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin, China
| | - Dong Wang
- Department of Neurosurgery, Tianjin Medical University, General Hospital, Tianjin, China.,Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin, China
| | - Wanqiang Su
- Department of Neurosurgery, Tianjin Medical University, General Hospital, Tianjin, China.,Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin, China
| | - Peng Wang
- Department of Neurosurgery, Tianjin Medical University, General Hospital, Tianjin, China.,Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin, China
| | - Linyue Guo
- Department of Neurosurgery, Tianjin Medical University, General Hospital, Tianjin, China.,Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin, China
| | - Wei Quan
- Department of Neurosurgery, Tianjin Medical University, General Hospital, Tianjin, China.,Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin, China
| | - Shuo An
- Department of Neurosurgery, Tianjin Medical University, General Hospital, Tianjin, China.,Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin, China
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University, General Hospital, Tianjin, China.,Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin, China
| | - Rongcai Jiang
- Department of Neurosurgery, Tianjin Medical University, General Hospital, Tianjin, China. .,Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin, China.
| |
Collapse
|