1
|
Hörauf JA, Schindler CR, Schaible I, Wang M, Weber B, El Saman A, Pallas C, Widera M, Marzi I, Henrich D, Leppik L. Extracellular vesicles epitopes as potential biomarker candidates in patients with traumatic spinal cord injury. Front Immunol 2024; 15:1478786. [PMID: 39703513 PMCID: PMC11656158 DOI: 10.3389/fimmu.2024.1478786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 11/06/2024] [Indexed: 12/21/2024] Open
Abstract
Background Extracellular vesicles (EVs), a heterogeneous group of cell-derived, membrane-enclosed vesicles bearing cell-specific epitopes, have been demonstrated to play a crucial role in neuronal-glial communication and the orchestration of neuroinflammatory processes. However, the existing evidence regarding their function as biomarkers and their role in the pathobiology of traumatic spinal cord injuries (tSCI), particularly in humans, is scarce. Objective The primary goal of this study was to investigate whether a distinct pattern of EV surface epitopes detected in the plasma of individuals suffering from spinal cord injury is indicative of tSCI. Methods The study includes patients with isolated tSCI (n=8), polytrauma patients without tSCI (PT; ISS ≥16, n=8), and healthy volunteers (HV; n=8). Plasma samples from tSCI and PT patients were collected right after admission to the emergency room (ER), 24 hours (24h), and 48h after trauma. EVs were isolated via size exclusion chromatography, and EVs' surface epitopes were quantified with MACSPlex EV Kit Neuro (prototype product, Miltenyi Biotec) and compared among the groups. Additionally, results were correlated with clinical parameters. Results In total, 19 epitopes differed significantly between the tSCI and the HV groups. Out of these 19, four (CD47, CD56, CD68, and ADAM17) were found to differ significantly among tSCI and PT groups. The expression of the CD47 epitope was found to correlate positively with the American Spinal Injury Association (ASIA) impairment scale. Conclusion We identified four potential EV-based tSCI biomarkers (CD47+, CD56+, CD68+, and ADAM17+ EVs) that differ in tSCI, with CD47+ EVs showing a strong correlation with the neurological function in tSCI. Thus, future studies might further specify the relevance of potential tSCI-specific biomarkers and investigate underlying mechanisms of tSCI.
Collapse
Affiliation(s)
- Jason-Alexander Hörauf
- Goethe University Frankfurt, University Hospital, Department of Trauma Surgery and Orthopedics, Frankfurt, Germany
| | - Cora Rebecca Schindler
- Goethe University Frankfurt, University Hospital, Department of Trauma Surgery and Orthopedics, Frankfurt, Germany
| | - Inna Schaible
- Goethe University Frankfurt, University Hospital, Department of Trauma Surgery and Orthopedics, Frankfurt, Germany
| | - Minhong Wang
- Goethe University Frankfurt, University Hospital, Department of Trauma Surgery and Orthopedics, Frankfurt, Germany
| | - Birte Weber
- Goethe University Frankfurt, University Hospital, Department of Trauma Surgery and Orthopedics, Frankfurt, Germany
| | - André El Saman
- Goethe University Frankfurt, University Hospital, Department of Trauma Surgery and Orthopedics, Frankfurt, Germany
| | - Christiane Pallas
- Goethe University Frankfurt, University Hospital, Institute for Medical Virology, Frankfurt, Germany
| | - Marek Widera
- Goethe University Frankfurt, University Hospital, Institute for Medical Virology, Frankfurt, Germany
| | - Ingo Marzi
- Goethe University Frankfurt, University Hospital, Department of Trauma Surgery and Orthopedics, Frankfurt, Germany
| | - Dirk Henrich
- Goethe University Frankfurt, University Hospital, Department of Trauma Surgery and Orthopedics, Frankfurt, Germany
| | - Liudmila Leppik
- Goethe University Frankfurt, University Hospital, Department of Trauma Surgery and Orthopedics, Frankfurt, Germany
| |
Collapse
|
2
|
Coenen H, Somers V, Fraussen J. Peripheral immune reactions following human traumatic spinal cord injury: the interplay of immune activation and suppression. Front Immunol 2024; 15:1495801. [PMID: 39664385 PMCID: PMC11631733 DOI: 10.3389/fimmu.2024.1495801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 11/05/2024] [Indexed: 12/13/2024] Open
Abstract
Traumatic spinal cord injury (SCI) damages the nerve tissue of the spinal cord, resulting in loss of motor and/or sensory functions at and below the injury level. SCI provokes a long-lasting immune response that extends beyond the spinal cord and induces changes in the composition and function of the peripheral immune system. Seemingly contradictory findings have been observed, as both systemic immune activation, including inflammation and autoimmunity, and immune suppression have been reported. Differences in the levels and functions of various cell types and components of both the innate and adaptive immune system supporting these changes have been described at (sub)acute and chronic stages post-injury. Further research is needed for a more comprehensive understanding of the peripheral immune reactions following SCI, their possible correlations with clinical characteristics, and how these immune responses could be targeted to facilitate the therapeutic management of SCI. In this review, we provide an overview of the current literature discussing changes in the peripheral immune system and their occurrence over time following a traumatic SCI.
Collapse
Affiliation(s)
| | | | - Judith Fraussen
- Department of Immunology and Infection, Biomedical Research Institute, UHasselt – Hasselt University, Hasselt, Belgium
| |
Collapse
|
3
|
Zhuo M, Deng Z, Yuan L, Mai Z, Zhong M, Ye JM. Association of systemic inflammatory response index and clinical outcome in acute traumatic spinal cord injury patients. Sci Rep 2024; 14:19085. [PMID: 39154138 PMCID: PMC11330529 DOI: 10.1038/s41598-024-69699-4] [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: 06/16/2024] [Accepted: 08/06/2024] [Indexed: 08/19/2024] Open
Abstract
Systemic inflammatory response index (SIRI) has been proven to be associated with the prognosis of coronary artery disease and many other diseases. However, the relationship between SIRI and acute traumatic spinal cord injury (tSCI) has rarely been evaluated. The study aims to assess the prognostic value of SIRI for clinical outcomes in individuals with acute tSCI. A total of 190 patients admitted within eight hours after tSCI between January 2021 and April 2023 were enrolled in our study. Logistic regression analysis was used to analyze the association between SIRI and American Spinal Injury Association Impairment Scale (AIS) grade at admission and discharge, as well as neurological improvement in tSCI patients, and receiver operating characteristic (ROC) analysis was performed to assess the discriminative ability of SIRI in predicting AIS grade at discharge. After adjusting for confounding factors, SIRI positively correlated with the AIS grade (A to C) at admission and discharge, and negatively correlated with neurological improvement. The area under the curve values in ROC analysis was 0.725 (95% CI 0.647, 0.803). The study suggests that SIRI is significantly associated with an increased risk of poor clinical outcome at discharge in tSCI patients and has a certain discriminative value.
Collapse
Affiliation(s)
- Ming Zhuo
- Medical College of Soochow University, Suzhou, 215123, Jiangsu, China
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Zihao Deng
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Lin Yuan
- Gannan Medical University, Ganzhou, 341000, China
| | - Zifeng Mai
- Gannan Medical University, Ganzhou, 341000, China
| | - Maolin Zhong
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Jun-Ming Ye
- Medical College of Soochow University, Suzhou, 215123, Jiangsu, China.
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China.
| |
Collapse
|
4
|
Smith D, Ridler M. Patient-reported outcome survey of user-experiences in the spinal cord injured-community with MPPT for treating wounds and pressure injuries and for controlling soft tissue infection caused by osteomyelitis. FRONTIERS IN REHABILITATION SCIENCES 2024; 5:1386518. [PMID: 38966822 PMCID: PMC11222669 DOI: 10.3389/fresc.2024.1386518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 06/10/2024] [Indexed: 07/06/2024]
Abstract
Background People with spinal cord injury (SCI) are at high risk of developing pressure injuries. Reports in the SCI-community had indicated that a new class of wound treatment, MPPT (micropore-particle-technology), was effective in treating pressure injuries. The British Spinal Injuries Association therefore conducted a survey among MPPT-users to learn from their experiences. Methods Online survey restricted to individuals with spinal cord injury. Participants were requested to identify themselves to permit validation of statement. Results The survey had 41 respondents reporting on a total of 49 wounds of which the two main categories were wounds (n = 33), primarily pelvic pressure ulcers; and draining fistulas (n = 9) caused by osteomyelitis. All wounds reported had reached full closure. Median duration of MPPT use and time to closure were 3 and 4 weeks for acute wounds (<6 weeks old) and 8 and 10 weeks for chronic wounds, respectively. On draining fistulas, MPPT had been used to reduce wound size, remove soft tissue infection, avoid sepsis, reduce autonomic dysreflexia, improve overall health, and avoid bed rest, whilst waiting for surgery. Comments on MPPT were 84% highly positive, 11% positive, and 0% negative. No adverse events were reported. Conclusions MPPT achieved a 100% closure rate of acute and chronic wounds, and, in draining fistulas, effectively controlled soft tissue infection resulting from osteomyelitis. MPPT does not require bed rest and is suitable for self-care and telemedicine, promoting independence and higher quality-of-life. The findings strongly agree with a recent clinical study of MPPT.
Collapse
Affiliation(s)
| | - Mark Ridler
- Spinal Injuries Association, Milton Keynes, United Kingdom
| |
Collapse
|
5
|
Song SS, Druschel LN, Conard JH, Wang JJ, Kasthuri NM, Ricky Chan E, Capadona JR. Depletion of complement factor 3 delays the neuroinflammatory response to intracortical microelectrodes. Brain Behav Immun 2024; 118:221-235. [PMID: 38458498 DOI: 10.1016/j.bbi.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/26/2024] [Accepted: 03/02/2024] [Indexed: 03/10/2024] Open
Abstract
The neuroinflammatory response to intracortical microelectrodes (IMEs) used with brain-machine interfacing (BMI) applications is regarded as the primary contributor to poor chronic performance. Recent developments in high-plex gene expression technologies have allowed for an evolution in the investigation of individual proteins or genes to be able to identify specific pathways of upregulated genes that may contribute to the neuroinflammatory response. Several key pathways that are upregulated following IME implantation are involved with the complement system. The complement system is part of the innate immune system involved in recognizing and eliminating pathogens - a significant contributor to the foreign body response against biomaterials. Specifically, we have identified Complement 3 (C3) as a gene of interest because it is the intersection of several key complement pathways. In this study, we investigated the role of C3 in the IME inflammatory response by comparing the neuroinflammatory gene expression at the microelectrode implant site between C3 knockout (C3-/-) and wild-type (WT) mice. We have found that, like in WT mice, implantation of intracortical microelectrodes in C3-/- mice yields a dramatic increase in the neuroinflammatory gene expression at all post-surgery time points investigated. However, compared to WT mice, C3 depletion showed reduced expression of many neuroinflammatory genes pre-surgery and 4 weeks post-surgery. Conversely, depletion of C3 increased the expression of many neuroinflammatory genes at 8 weeks and 16 weeks post-surgery, compared to WT mice. Our results suggest that C3 depletion may be a promising therapeutic target for acute, but not chronic, relief of the neuroinflammatory response to IME implantation. Additional compensatory targets may also be required for comprehensive long-term reduction of the neuroinflammatory response for improved intracortical microelectrode performance.
Collapse
Affiliation(s)
- Sydney S Song
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, United States.
| | - Lindsey N Druschel
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, United States.
| | - Jacob H Conard
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States.
| | - Jaime J Wang
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, United States.
| | - Niveda M Kasthuri
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, United States.
| | - E Ricky Chan
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH 44106, United States.
| | - Jeffrey R Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, United States.
| |
Collapse
|
6
|
Yang P, Bian ZQ, Song ZB, Yang CY, Wang L, Yao ZX. Dominant mechanism in spinal cord injury-induced immunodeficiency syndrome (SCI-IDS): sympathetic hyperreflexia. Rev Neurosci 2024; 35:259-269. [PMID: 37889575 DOI: 10.1515/revneuro-2023-0090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023]
Abstract
Clinical studies have shown that individuals with spinal cord injury (SCI) are particularly susceptible to infectious diseases, resulting in a syndrome called SCI-induced immunodeficiency syndrome (SCI-IDS), which is the leading cause of death after SCI. It is believed that SCI-IDS is associated with exaggerated activation of sympathetic preganglionic neurons (SPNs). After SCI, disruption of bulbospinal projections from the medulla oblongata C1 neurons to the SPNs results in the loss of sympathetic inhibitory modulation from the brain and brainstem and the occurrence of abnormally high levels of spinal sympathetic reflexes (SSR), named sympathetic hyperreflexia. As the post-injury survival time lengthens, mass recruitment and anomalous sprouting of excitatory interneurons within the spinal cord result in increased SSR excitability, resulting in an excess sympathetic output that disrupts the immune response. Therefore, we first analyze the structural underpinnings of the spinal cord-sympathetic nervous system-immune system after SCI, then demonstrate the progress in highlighting mechanisms of SCI-IDS focusing on norepinephrine (NE)/Beta 2-adrenergic receptor (β2-AR) signal pathways, and summarize recent preclinical studies examining potential means such as regulating SSR and inhibiting β2-AR signal pathways to improve immune function after SCI. Finally, we present research perspectives such as to promote the effective regeneration of C1 neurons to rebuild the connection of C1 neurons with SPNs, to regulate excitable or inhibitory interneurons, and specifically to target β2-AR signal pathways to re-establish neuroimmune balance. These will help us design effective strategies to reverse post-SCI sympathetic hyperreflexia and improve the overall quality of life for individuals with SCI.
Collapse
Affiliation(s)
- Ping Yang
- Department of Neurobiology, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Zhi-Qun Bian
- Department of Orthopedics, The Second Affiliated Hospital of Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Zhen-Bo Song
- Department of Physiology, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Cheng-Ying Yang
- Department of Immunology, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Li Wang
- Department of Immunology, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Zhong-Xiang Yao
- Department of Physiology, Army Medical University (Third Military Medical University), Chongqing 400038, China
| |
Collapse
|
7
|
Nekanti U, Sakthivel PS, Zahedi A, Creasman DA, Nishi RA, Dumont CM, Piltti KM, Guardamondo GL, Hernandez N, Chen X, Song H, Lin X, Martinez J, On L, Lakatos A, Pawar K, David BT, Guo Z, Seidlits SK, Xu X, Shea LD, Cummings BJ, Anderson AJ. Multichannel bridges and NSC synergize to enhance axon regeneration, myelination, synaptic reconnection, and recovery after SCI. NPJ Regen Med 2024; 9:12. [PMID: 38499577 PMCID: PMC10948859 DOI: 10.1038/s41536-024-00356-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 02/15/2024] [Indexed: 03/20/2024] Open
Abstract
Regeneration in the injured spinal cord is limited by physical and chemical barriers. Acute implantation of a multichannel poly(lactide-co-glycolide) (PLG) bridge mechanically stabilizes the injury, modulates inflammation, and provides a permissive environment for rapid cellularization and robust axonal regrowth through this otherwise inhibitory milieu. However, without additional intervention, regenerated axons remain largely unmyelinated (<10%), limiting functional repair. While transplanted human neural stem cells (hNSC) myelinate axons after spinal cord injury (SCI), hNSC fate is highly influenced by the SCI inflammatory microenvironment, also limiting functional repair. Accordingly, we investigated the combination of PLG scaffold bridges with hNSC to improve histological and functional outcome after SCI. In vitro, hNSC culture on a PLG scaffold increased oligodendroglial lineage selection after inflammatory challenge. In vivo, acute PLG bridge implantation followed by chronic hNSC transplantation demonstrated a robust capacity of donor human cells to migrate into PLG bridge channels along regenerating axons and integrate into the host spinal cord as myelinating oligodendrocytes and synaptically integrated neurons. Axons that regenerated through the PLG bridge formed synaptic circuits that connected the ipsilateral forelimb muscle to contralateral motor cortex. hNSC transplantation significantly enhanced the total number of regenerating and myelinated axons identified within the PLG bridge. Finally, the combination of acute bridge implantation and hNSC transplantation exhibited robust improvement in locomotor recovery. These data identify a successful strategy to enhance neurorepair through a temporally layered approach using acute bridge implantation and chronic cell transplantation to spare tissue, promote regeneration, and maximize the function of new axonal connections.
Collapse
Affiliation(s)
- Usha Nekanti
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA.
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA.
| | - Pooja S Sakthivel
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
| | - Atena Zahedi
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Dana A Creasman
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
| | - Rebecca A Nishi
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Courtney M Dumont
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Katja M Piltti
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Glenn L Guardamondo
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Norbert Hernandez
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Xingyuan Chen
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Hui Song
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Xiaoxiao Lin
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
| | - Joshua Martinez
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Lillian On
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Anita Lakatos
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Kiran Pawar
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Brian T David
- Department of Neurosurgery, Rush University Medical Center, Chicago, IL, USA
| | - Zhiling Guo
- Department of Medicine & Susan Samueli Integrative Health Institute, University of California, Irvine, CA, USA
| | - Stephanie K Seidlits
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
- Center for Neural Circuit Mapping, University of California Irvine, Irvine, CA, USA
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Brian J Cummings
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
- Department of Physical Medicine and Rehabilitation, University of California, Irvine, CA, USA
- Institute for Memory Impairments & Neurological Disorder, University of California Irvine, Irvine, CA, USA
| | - Aileen J Anderson
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA.
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA.
- Department of Physical Medicine and Rehabilitation, University of California, Irvine, CA, USA.
- Institute for Memory Impairments & Neurological Disorder, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
8
|
Guntermann A, Marcus K, May C. The good or the bad: an overview of autoantibodies in traumatic spinal cord injury. Biol Chem 2024; 405:79-89. [PMID: 37786927 DOI: 10.1515/hsz-2023-0252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/12/2023] [Indexed: 10/04/2023]
Abstract
Infections remain the most common cause of death after traumatic spinal cord injury, likely due to a developing immune deficiency syndrome. This, together with a somewhat contradictory development of autoimmunity in many patients, are two major components of the maladaptive systemic immune response. Although the local non-resolving inflammation in the lesioned spinal cord may lead to an antibody formation against autoantigens of the injured spinal cord tissue, there are also natural (pre-existing) autoantibodies independent of the injury. The way in which these autoantibodies with different origins affect the neuronal and functional outcome of spinal cord-injured patients is still controversial.
Collapse
Affiliation(s)
- Annika Guntermann
- Medical Proteome Analysis, Center for Protein Diagnostics (ProDi), Ruhr University Bochum, D-44801 Bochum, Germany
- Medizinisches Proteom-Center, Medical Faculty, ProDi E2.233, Ruhr University Bochum, Gesundheitscampus 4, D-44801 Bochum, Germany
| | - Katrin Marcus
- Medical Proteome Analysis, Center for Protein Diagnostics (ProDi), Ruhr University Bochum, D-44801 Bochum, Germany
- Medizinisches Proteom-Center, Medical Faculty, ProDi E2.233, Ruhr University Bochum, Gesundheitscampus 4, D-44801 Bochum, Germany
| | - Caroline May
- Medical Proteome Analysis, Center for Protein Diagnostics (ProDi), Ruhr University Bochum, D-44801 Bochum, Germany
- Medizinisches Proteom-Center, Medical Faculty, ProDi E2.233, Ruhr University Bochum, Gesundheitscampus 4, D-44801 Bochum, Germany
| |
Collapse
|
9
|
Sams-Dodd J, Belci M, Bandi S, Smith D, Sams-Dodd F. Stable closure of acute and chronic wounds and pressure ulcers and control of draining fistulas from osteomyelitis in persons with spinal cord injuries: non-interventional study of MPPT passive immunotherapy delivered via telemedicine in community care. Front Med (Lausanne) 2024; 10:1279100. [PMID: 38249963 PMCID: PMC10797031 DOI: 10.3389/fmed.2023.1279100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 12/08/2023] [Indexed: 01/23/2024] Open
Abstract
Background Micropore particle technology (MPPT) is a topical wound treatment. It is a passive immunotherapy, acting via the skin and wound microbiome without the use of antimicrobial action. In a general patient population, it removed wound infections 60% and initiated tissue regeneration 50% quicker than antibiotics and antiseptics. As MPPT supports the immune system, the aim was to confirm that MPPT is also effective in immunocompromised individuals. People with spinal cord injury (SCI) are immunodeficient due to their injury and not an underlying disease and recruit 50% fewer immune cells to an injury. The study, therefore, determined the efficacy, safety, health economics, and sustainability of MPPT in acute and chronic wounds and pressure ulcers in this patient population. Methods Pressure ulcers in SCI persons are an orphan indication, patient variability is high, and ICH E10 excludes comparators due to ethical concerns. The study design was, therefore, a single-arm, non-interventional, observational, post-market surveillance study of MPPT for treating wounds and pressure ulcers and removing soft tissue infection in connection with draining fistulas in SCI persons. The study was based on telemedicine in community care. Results The study included 44 wounds. All acute and chronic grade 1-4 wounds and pressure ulcers reached stable closure. In wounds acting as fistulas draining from an underlying, primary focus of infection, e.g., osteomyelitis, MPPT removed the soft tissue infection in approx. 2.5 months and supported regeneration, considerably reducing fistula sizes. Compared to standard care, per-wound cost savings were 51 to 94% depending on wound grade and age, and substantial nursing resources were freed up. The telemedicine approach was well received by participants and supported independence and self-care. The use of antimicrobials, plastics, and synthetic polymers was essentially eliminated. MPPT did not require bed rest. Conclusion The study confirmed that MPPT is safe and effective in treating acute and chronic wounds in immunocompetent and immunocompromised individuals, including wounds with antimicrobial-resistant infections. MPPT also removes soft tissue infections caused by an underlying primary focus of infection, such as osteomyelitis. Non-healing wounds currently represent an unmet clinical need. The findings suggest that a therapy acting via the microbiome without antimicrobial actions is effective.
Collapse
Affiliation(s)
| | - Maurizio Belci
- The National Spinal Injuries Centre, Stoke Mandeville Hospital, Aylesbury, Buckinghamshire, United Kingdom
| | - Surendra Bandi
- Duke of Cornwall Spinal Treatment Centre, Salisbury District Hospital, Salisbury, Wiltshire, United Kingdom
| | - Damian Smith
- Duke of Cornwall Spinal Treatment Centre, Salisbury District Hospital, Salisbury, Wiltshire, United Kingdom
| | | |
Collapse
|
10
|
Morrison D, Pinpin C, Lee A, Sison C, Chory A, Gregersen PK, Forrest G, Kirshblum S, Harkema SJ, Boakye M, Harrop JS, Bryce TN, Schwab JM, Kwon BK, Stein AB, Bank MA, Bloom O. Profiling Immunological Phenotypes in Individuals During the First Year After Traumatic Spinal Cord Injury: A Longitudinal Analysis. J Neurotrauma 2023; 40:2621-2637. [PMID: 37221869 PMCID: PMC10722895 DOI: 10.1089/neu.2022.0500] [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] [Indexed: 05/25/2023] Open
Abstract
Abstract Individuals with SCI are severely affected by immune system changes, resulting in increased risk of infections and persistent systemic inflammation. While recent data support that immunological changes after SCI differ in the acute and chronic phases of living with SCI, only limited immunological phenotyping in humans is available. To characterize dynamic molecular and cellular immune phenotypes over the first year, we assess RNA (bulk-RNA sequencing), protein, and flow cytometry (FACS) profiles of blood samples from 12 individuals with SCI at 0-3 days and at 3, 6, and 12 months post injury (MPI) compared to 23 uninjured individuals (controls). We identified 967 differentially expressed (DE) genes in individuals with SCI (FDR <0.001) compared to controls. Within the first 6 MPI we detected a reduced expression of NK cell genes, consistent with reduced frequencies of CD56bright, CD56dim NK cells present at 12 MPI. Over 6MPI, we observed increased and prolonged expression of genes associated with inflammation (e.g. HMGB1, Toll-like receptor signaling) and expanded frequencies of monocytes acutely. Canonical T-cell related DE genes (e.g. FOXP3, TCF7, CD4) were upregulated during the first 6 MPI and increased frequencies of activated T cells at 3-12 MPI. Neurological injury severity was reflected in distinct whole blood gene expression profiles at any time after SCI, verifying a persistent 'neurogenic' imprint. Overall, 2876 DE genes emerge when comparing motor complete to motor incomplete SCI (ANOVA, FDR <0.05), including those related to neutrophils, inflammation, and infection. In summary, we identify a dynamic immunological phenotype in humans, including molecular and cellular changes which may provide potential targets to reduce inflammation, improve immunity, or serve as candidate biomarkers of injury severity.
Collapse
Affiliation(s)
- Debra Morrison
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA
| | - Camille Pinpin
- Donald and Barbara Zucker School of Medicine at Hofstra Northwell, Hempstead, New York, USA
| | - Annette Lee
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA
- Donald and Barbara Zucker School of Medicine at Hofstra Northwell, Hempstead, New York, USA
| | - Cristina Sison
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA
- Donald and Barbara Zucker School of Medicine at Hofstra Northwell, Hempstead, New York, USA
| | - Ashley Chory
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA
| | - Peter K. Gregersen
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA
- Donald and Barbara Zucker School of Medicine at Hofstra Northwell, Hempstead, New York, USA
| | - Gail Forrest
- Tim and Caroline Reynolds Center for Spinal Stimulation, Center for Mobility and Human Engineering Research, West Orange, New Jersey, USA
- Department of Physical Medicine and Rehabilitation, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Steven Kirshblum
- Tim and Caroline Reynolds Center for Spinal Stimulation, Center for Mobility and Human Engineering Research, West Orange, New Jersey, USA
- Department of Physical Medicine and Rehabilitation, Rutgers New Jersey Medical School, Newark, New Jersey, USA
- Kessler Institute for Rehabilitation. West Orange, New Jersey, USA
| | - Susan J. Harkema
- Kentucky Spinal Injury Research Center, School of Medicine, University of Louisville, Louisville, Kentucky, USA
- Department of Neurosurgery, School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Maxwell Boakye
- Kentucky Spinal Injury Research Center, School of Medicine, University of Louisville, Louisville, Kentucky, USA
- Department of Neurosurgery, School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - James S. Harrop
- Department of Neurosurgery, Thomas Jefferson University Hospitals, Philadelphia, Pennsylvania, USA
| | - Thomas N. Bryce
- Department of Rehabilitation and Human Performance, Icahn School of Medicine at Mount Sinai Hospital, New York, New York, USA
| | - Jan M. Schwab
- The Belford Center for Spinal Cord Injury, Spinal Cord Division, Wexner Medical Center, The Ohio State University, Columbus, Ohio, USA
- Department of Neurology, Spinal Cord Division, Wexner Medical Center, The Ohio State University, Columbus, Ohio, USA
| | - Brian K. Kwon
- International Collaboration on Repair Discoveries (ICORD), Department of Orthopaedics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Adam B. Stein
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA
- Donald and Barbara Zucker School of Medicine at Hofstra Northwell, Hempstead, New York, USA
| | - Matthew A. Bank
- Donald and Barbara Zucker School of Medicine at Hofstra Northwell, Hempstead, New York, USA
- North Shore University Hospital, Manhasset, New York, USA
| | - Ona Bloom
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA
- Donald and Barbara Zucker School of Medicine at Hofstra Northwell, Hempstead, New York, USA
| |
Collapse
|
11
|
Song S, Druschel LN, Chan ER, Capadona JR. Differential expression of genes involved in the chronic response to intracortical microelectrodes. Acta Biomater 2023; 169:348-362. [PMID: 37507031 PMCID: PMC10528922 DOI: 10.1016/j.actbio.2023.07.038] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 07/13/2023] [Accepted: 07/23/2023] [Indexed: 07/30/2023]
Abstract
Brain-Machine Interface systems (BMIs) are clinically valuable devices that can provide functional restoration for patients with spinal cord injury or improved integration for patients requiring prostheses. Intracortical microelectrodes can record neuronal action potentials at a resolution necessary for precisely controlling BMIs. However, intracortical microelectrodes have a demonstrated history of progressive decline in the recording performance with time, inhibiting their usefulness. One major contributor to decreased performance is the neuroinflammatory response to the implanted microelectrodes. The neuroinflammatory response can lead to neurodegeneration and the formation of a glial scar at the implant site. Historically, histological imaging of relatively few known cellular and protein markers has characterized the neuroinflammatory response to implanted microelectrode arrays. However, neuroinflammation requires many molecular players to coordinate the response - meaning traditional methods could result in an incomplete understanding. Taking advantage of recent advancements in tools to characterize the relative or absolute DNA/RNA expression levels, a few groups have begun to explore gene expression at the microelectrode-tissue interface. We have utilized a custom panel of ∼813 neuroinflammatory-specific genes developed with NanoString for bulk tissue analysis at the microelectrode-tissue interface. Our previous studies characterized the acute innate immune response to intracortical microelectrodes. Here we investigated the gene expression at the microelectrode-tissue interface in wild-type (WT) mice chronically implanted with nonfunctioning probes. We found 28 differentially expressed genes at chronic time points (4WK, 8WK, and 16WK), many in the complement and extracellular matrix system. Further, the expression levels were relatively stable over time. Genes identified here represent chronic molecular players at the microelectrode implant sites and potential therapeutic targets for the long-term integration of microelectrodes. STATEMENT OF SIGNIFICANCE: Intracortical microelectrodes can record neuronal action potentials at a resolution necessary for the precise control of Brain-Machine Interface systems (BMIs). However, intracortical microelectrodes have a demonstrated history of progressive declines in the recording performance with time, inhibiting their usefulness. One major contributor to the decline in these devices is the neuroinflammatory response against the implanted microelectrodes. Historically, neuroinflammation to implanted microelectrode arrays has been characterized by histological imaging of relatively few known cellular and protein markers. Few studies have begun to develop a more in-depth understanding of the molecular pathways facilitating device-mediated neuroinflammation. Here, we are among the first to identify genetic pathways that could represent targets to improve the host response to intracortical microelectrodes, and ultimately device performance.
Collapse
Affiliation(s)
- Sydney Song
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, United States
| | - Lindsey N Druschel
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, United States
| | - E Ricky Chan
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Jeffrey R Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, United States.
| |
Collapse
|
12
|
Michael FM, Patel SP, Bachstetter AD, Rabchevsky AG. Proinflammatory and Immunomodulatory Gene and Protein Expression Patterns in Spinal Cord and Spleen Following Acute and Chronic High Thoracic Injury. J Inflamm Res 2023; 16:3341-3349. [PMID: 37576153 PMCID: PMC10423003 DOI: 10.2147/jir.s417435] [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] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 08/04/2023] [Indexed: 08/15/2023] Open
Abstract
Introduction In addition to paralysis and loss of sensation, high-level spinal cord injury (SCI) causes sympathetic dysfunction that can lead to autonomic dysreflexia (AD) and chronic immune suppression involving splenic leukopenia. Evidence has shown that treatment with either gabapentin or blockade of TNFα mitigates maladaptive plasticity and the underlying hemodynamic dysfunction, spleen atrophy, and immune dysfunction associated with AD. Because significant improvements long term was noted following treatments only during acute stages of recovery, we sought to systematically examine changes in proinflammatory and immunomodulatory cytokines to ascertain the reason. Methods Adult female Wistar rats underwent complete T4 spinal transection before euthanasia at systematic intervals from 3 days to 8 weeks after injury. Using qRT-PCR and meso scale discovery (MSD) assays, the gene and protein expression of TNFα and IFNγ in the spleen, upper thoracic (T4-9) and lumbosacral (L5-S6) spinal cords were analyzed. Results We found that spleen atrophy occurs in a biphasic manner compared to naïve controls, with significant decreases in the spleen mass noted at 3 days and 8 weeks after injury. Splenic TNFα mRNA and protein levels did not change significantly over time, while IFNγ gene expression dipped acutely with trends for increased protein levels at more chronic time points. TNFα protein increased significantly only in thoracic spinal cord segments from 3 to 14 days post-injury. IFNγ mRNA and protein levels remained unelevated in injured spinal cords over time, with trends for increased protein levels at 2 and 8 weeks in the lumbosacral segments. Discussion Novel temporal-spatial cytokine expression profiles reveal that TNFα protein levels are increased solely in upper thoracic segments after high thoracic SCI, while IFNγ remains unaltered. Splenic leukopenia and latent systemic immunosuppression are not associated with altered TNFα or IFNγ expression in the spleen or spinal cord.
Collapse
Affiliation(s)
- Felicia M Michael
- Department of Physiology, University of Kentucky, Lexington, KY, 40536-0509, USA
- Spinal Cord & Brain Injury Research Center (SCoBIRC); University of Kentucky, Lexington, KY, 40536-0509, USA
| | - Samir P Patel
- Department of Physiology, University of Kentucky, Lexington, KY, 40536-0509, USA
- Spinal Cord & Brain Injury Research Center (SCoBIRC); University of Kentucky, Lexington, KY, 40536-0509, USA
| | - Adam D Bachstetter
- Spinal Cord & Brain Injury Research Center (SCoBIRC); University of Kentucky, Lexington, KY, 40536-0509, USA
- Department of Neuroscience, University of Kentucky, Lexington, KY, 40536-0509, USA
| | - Alexander G Rabchevsky
- Department of Physiology, University of Kentucky, Lexington, KY, 40536-0509, USA
- Spinal Cord & Brain Injury Research Center (SCoBIRC); University of Kentucky, Lexington, KY, 40536-0509, USA
| |
Collapse
|
13
|
Ortega MA, Fraile-Martinez O, García-Montero C, Haro S, Álvarez-Mon MÁ, De Leon-Oliva D, Gomez-Lahoz AM, Monserrat J, Atienza-Pérez M, Díaz D, Lopez-Dolado E, Álvarez-Mon M. A comprehensive look at the psychoneuroimmunoendocrinology of spinal cord injury and its progression: mechanisms and clinical opportunities. Mil Med Res 2023; 10:26. [PMID: 37291666 PMCID: PMC10251601 DOI: 10.1186/s40779-023-00461-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 06/01/2023] [Indexed: 06/10/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating and disabling medical condition generally caused by a traumatic event (primary injury). This initial trauma is accompanied by a set of biological mechanisms directed to ameliorate neural damage but also exacerbate initial damage (secondary injury). The alterations that occur in the spinal cord have not only local but also systemic consequences and virtually all organs and tissues of the body incur important changes after SCI, explaining the progression and detrimental consequences related to this condition. Psychoneuroimmunoendocrinology (PNIE) is a growing area of research aiming to integrate and explore the interactions among the different systems that compose the human organism, considering the mind and the body as a whole. The initial traumatic event and the consequent neurological disruption trigger immune, endocrine, and multisystem dysfunction, which in turn affect the patient's psyche and well-being. In the present review, we will explore the most important local and systemic consequences of SCI from a PNIE perspective, defining the changes occurring in each system and how all these mechanisms are interconnected. Finally, potential clinical approaches derived from this knowledge will also be collectively presented with the aim to develop integrative therapies to maximize the clinical management of these patients.
Collapse
Affiliation(s)
- Miguel A. Ortega
- Department of Medicine and Medical Specialities, University of Alcala, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities, University of Alcala, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Cielo García-Montero
- Department of Medicine and Medical Specialities, University of Alcala, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Sergio Haro
- Department of Medicine and Medical Specialities, University of Alcala, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Miguel Ángel Álvarez-Mon
- Department of Medicine and Medical Specialities, University of Alcala, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Department of Psychiatry and Mental Health, Hospital Universitario Infanta Leonor, 28031 Madrid, Spain
| | - Diego De Leon-Oliva
- Department of Medicine and Medical Specialities, University of Alcala, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Ana M. Gomez-Lahoz
- Department of Medicine and Medical Specialities, University of Alcala, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Jorge Monserrat
- Department of Medicine and Medical Specialities, University of Alcala, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Mar Atienza-Pérez
- Service of Rehabilitation, National Hospital for Paraplegic Patients, Carr. de la Peraleda, S/N, 45004 Toledo, Spain
| | - David Díaz
- Department of Medicine and Medical Specialities, University of Alcala, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Elisa Lopez-Dolado
- Department of Medicine and Medical Specialities, University of Alcala, 28801 Alcala de Henares, Spain
- Department of Psychiatry and Mental Health, Hospital Universitario Infanta Leonor, 28031 Madrid, Spain
| | - Melchor Álvarez-Mon
- Department of Medicine and Medical Specialities, University of Alcala, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Immune System Diseases-Rheumatology Service and Internal Medicine, University Hospital Príncipe de Asturias (CIBEREHD), 28806 Alcala de Henares, Spain
| |
Collapse
|
14
|
Kartal O, Yilmaz B, Gülec M, Guzelkucuk U, Kenan Tan A, Sener O, Muşabak U. Compromised T-cell immunity in patients with spinal cord injury and its relationship with injury characteristics. Turk J Phys Med Rehabil 2023; 69:69-74. [PMID: 37201015 PMCID: PMC10186008 DOI: 10.5606/tftrd.2023.11658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 12/20/2022] [Indexed: 05/20/2023] Open
Abstract
Objectives The aim of this study was to investigate in vivo and in vitro cellular immune responses in patients with chronic (spinal cord injury; SCI), determine the effects of autonomic dysfunction on cellular immune response, and determine the effect of completeness of the injury at different levels on cellular immune response. Patients and methods Forty-nine patients (42 males, 7 females; mean age: 35.5±13.4 years; range, 18 to 68 years) with chronic (time since injury >6 months) traumatic SCI were included in this cross sectional study between March 2013 and December 2013. Patients were allocated into two groups: Group 1, patients with an injury at T7 or below, and Group 2, patients with an injury at T6 or above. All patients in Group 2 had a history of autonomic dysreflexia and orthostatic hypotension. Intradermal skin tests were applied to the participants to reveal delayed T-cell responses. The percentages of cluster of differentiation (CD)3+ T cells and CD3+ T cells expressing CD69 and CD25 were analyzed by flow cytometry for the detection of activated T cells including all T-cell subsets. Results When patients with complete injuries were compared, the CD45+ cell percentage was found to be significantly higher in patients in Group 2. Patients with an incomplete SCI had increased skin response to candida antigens compared to complete SCI patients. Incomplete SCI patients also had higher percentages of lymphocytes and CD3+CD25+ and CD3+CD69+ T cells compared to patients with complete SCI. Conclusion T-cell activity is impaired in chronic SCI patients with higher levels of injury, and the completeness of injury and autonomic dysfunction gain prominence as compromising factors in T-cell immunity.
Collapse
Affiliation(s)
- Ozgur Kartal
- Department of Immunology and Allergic Diseases, Gülhane Faculty of Medicine and Gülhane Training and Research Hospital, University of Health Sciences, Ankara, Türkiye
| | - Bilge Yilmaz
- Department of Physical Medicine and Rehabilitation, University of Health Sciences, Ankara Gaziler Physical Medicine and Rehabilitation Training and Research Hospital, Ankara, Türkiye
| | - Mustafa Gülec
- Department of Immunology and Allergic Diseases, Private Güven Hospital, Ankara, Türkiye
| | - Umüt Guzelkucuk
- Department of Physical Medicine and Rehabilitation, University of Health Sciences, Ankara Gaziler Physical Medicine and Rehabilitation Training and Research Hospital, Ankara, Türkiye
| | - Arif Kenan Tan
- Department of Physical Medicine and Rehabilitation, University of Health Sciences, Ankara Gaziler Physical Medicine and Rehabilitation Training and Research Hospital, Ankara, Türkiye
| | - Osman Sener
- Department of Immunology and Allergic Diseases, Gülhane Faculty of Medicine and Gülhane Training and Research Hospital, University of Health Sciences, Ankara, Türkiye
| | - Uğur Muşabak
- Department of Immunology and Allergic Diseases, Baskent University, Faculty of Medicine, Ankara, Türkiye
| |
Collapse
|
15
|
Davis M, Jethani L, Robbins E, Kaner M. Is It Really the Foley? A Systematic Review of Bladder Management and Infection Risk. Top Spinal Cord Inj Rehabil 2023; 29:94-107. [PMID: 36819923 PMCID: PMC9936901 DOI: 10.46292/sci22-00009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Background The belief that intermittent catheterization results in fewer infections than indwelling catheters is commonly expressed in the spinal cord injury literature. Some practice guidelines strongly recommend intermittent over indwelling catheterization due to concerns about infections and other complications. However, studies on this topic are of low quality. Guidelines from the Consortium for Spinal Cord Medicine suggest the data regarding infection risk are mixed, and they do not recommend one bladder management method over the other. Objectives To compare risk of bias in studies reporting higher rates of urinary tract infection (UTI) with indwelling catheters to studies that found equal rates of UTI between indwelling and intermittent catheterization, and to describe implications in clinical decision-making. Methods A systematic search of PubMed, CINAHL, Embase, and SCOPUS databases from January 1, 1980, to September 15, 2020, was conducted. Eligible studies compared symptomatic UTI rates between indwelling and intermittent catheterization. We used a risk of bias assessment tool to evaluate each study. Results Twenty-four studies were identified. Only three of these reported significantly higher UTI risk with indwelling catheters, and all three demonstrated a critical risk of bias. More than half of the studies reported differences in UTI risk of less than 20% between the two methods. Studies with larger (nonsignificant) differences favoring intermittent catheterization were more susceptible to bias from confounding. Conclusion The hypothesis that indwelling catheters cause more UTIs than intermittent catheterization is not supported by the scientific literature. Most studies failed to demonstrate a significant difference in UTI risk, and studies with nonsignificant trends favoring intermittent catheterization were more susceptible to bias from confounding. Perceived risk of infection should not influence a patient's choice of catheter type.
Collapse
Affiliation(s)
- Matthew Davis
- Department of Physical Medicine and Rehabilitation, McGovern Medical School, Houston, Texas
| | - Lavina Jethani
- Department of Physical Medicine and Rehabilitation, McGovern Medical School, Houston, Texas
| | - Emily Robbins
- Department of Physical Medicine and Rehabilitation, McGovern Medical School, Houston, Texas
| | - Mahmut Kaner
- Department of Physical Medicine and Rehabilitation, McGovern Medical School, Houston, Texas
| |
Collapse
|
16
|
Ayala C, Fishman M, Noyelle M, Bassiri H, Young W. Species Differences in Blood Lymphocyte Responses After Spinal Cord Injury. J Neurotrauma 2023; 40:807-819. [PMID: 36367185 PMCID: PMC10150731 DOI: 10.1089/neu.2022.0122] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
People with spinal cord injury (SCI) get recurrent infections, such as urinary tract infections (UTIs) and pneumonias, that cause mortality and worsen neurological recovery. Over the past decades, researchers have proposed that post-SCI lymphopenia and decreased lymphocyte function increase susceptibility to infections and worsen neurological outcome in humans, leading to a condition called SCI-induced immune depression syndrome (SCI-IDS). In this review, we explore how SCI affects blood lymphocyte homeostasis and function in humans and rodents. Understanding how SCI affects blood lymphocytes will help the management of recurrent infections in spinal cord injured people and shed light on the clinical translation of findings in animal models to humans.
Collapse
Affiliation(s)
- Carlos Ayala
- W.M. Keck Center for Collaborative Neuroscience, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA.,New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | - Morgan Fishman
- W.M. Keck Center for Collaborative Neuroscience, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Margot Noyelle
- W.M. Keck Center for Collaborative Neuroscience, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Hamid Bassiri
- Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Wise Young
- W.M. Keck Center for Collaborative Neuroscience, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| |
Collapse
|
17
|
Grassner L, Klein B, Garcia-Ovejero D, Mach O, Scheiblhofer S, Weiss R, Vargas-Baquero E, Kramer JLK, Leister I, Rohde E, Oeller M, Molina-Holgado E, Griessenauer CJ, Maier D, Aigner L, Arevalo-Martin A. Systemic Immune Profile Predicts the Development of Infections in Patients with Spinal Cord Injuries. J Neurotrauma 2022; 39:1678-1686. [PMID: 35607859 DOI: 10.1089/neu.2021.0448] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Patients with spinal cord injury (SCI) frequently develop infections that may affect quality of life, be life-threatening, and impair their neurological recovery in the acute and subacute injury phases. Therefore, identifying patients with SCI at risk for developing infections in this stage is of utmost importance. We determined the systemic levels of immune cell populations, cytokines, chemokines, and growth factors in 81 patients with traumatic SCI at 4 weeks after injury and compared them with those of 26 age-matched healthy control subjects. Patients who developed infections between 4 and 16 weeks after injury exhibited higher numbers of neutrophils and eosinophils, as well as lower numbers of lymphocytes and eotaxin-1 (CCL11) levels. Accordingly, lasso logistic regression showed that incomplete lesions (American Spinal Injury Association Impairment Scale [AIS] C and D grades), the levels of eotaxin-1, and the number of lymphocytes, basophils, and monocytes are predictive of lower odds for infections. On the other hand, the number of neutrophils and eosinophils as well as, in a lesser extent, the levels of IP-10 (CXCL10), MCP-1 (CCL2), BDNF [brain-derived neurotrophic factor], and vascular endothelial growth factor [VEGF]-A, are predictors of increased susceptibility for developing infections. Overall, our results point to systemic immune disbalance after SCI as predictors of infection in a period when infections may greatly interfere with neurological and functional recovery and suggest new pathways and players to further explore novel therapeutic strategies.
Collapse
Affiliation(s)
- Lukas Grassner
- Institute of Molecular Regenerative Medicine, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria.,ParaMove, SCI Research Unit, BG Trauma Center Murnau, Murnau, Germany, and Paracelsus Medical University, Salzburg, Austria.,Spinal Cord Injury Center, BG Trauma Center Murnau, Murnau, Germany
| | - Barbara Klein
- Institute of Molecular Regenerative Medicine, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Daniel Garcia-Ovejero
- Laboratory of Neuroinflammation, Hospital Nacional de Paraplejicos, SESCAM, Toledo, Spain
| | - Orpheus Mach
- ParaMove, SCI Research Unit, BG Trauma Center Murnau, Murnau, Germany, and Paracelsus Medical University, Salzburg, Austria.,Spinal Cord Injury Center, BG Trauma Center Murnau, Murnau, Germany
| | - Sandra Scheiblhofer
- Division of Allergy and Immunology, Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | - Richard Weiss
- Division of Allergy and Immunology, Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | | | - John L K Kramer
- International Collaboration on Repair Discoveries (ICORD), Pharmacology, and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Iris Leister
- Institute of Molecular Regenerative Medicine, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria.,ParaMove, SCI Research Unit, BG Trauma Center Murnau, Murnau, Germany, and Paracelsus Medical University, Salzburg, Austria.,Spinal Cord Injury Center, BG Trauma Center Murnau, Murnau, Germany
| | - Eva Rohde
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria.,Department for Transfusion Medicine, University Hospital of Salzburg (SALK), University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Michaela Oeller
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria.,Department for Transfusion Medicine, University Hospital of Salzburg (SALK), University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Eduardo Molina-Holgado
- Laboratory of Neuroinflammation, Hospital Nacional de Paraplejicos, SESCAM, Toledo, Spain
| | - Christoph J Griessenauer
- Department of Neurosurgery, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Doris Maier
- ParaMove, SCI Research Unit, BG Trauma Center Murnau, Murnau, Germany, and Paracelsus Medical University, Salzburg, Austria.,Spinal Cord Injury Center, BG Trauma Center Murnau, Murnau, Germany
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria.,ParaMove, SCI Research Unit, BG Trauma Center Murnau, Murnau, Germany, and Paracelsus Medical University, Salzburg, Austria
| | - Angel Arevalo-Martin
- Laboratory of Neuroinflammation, Hospital Nacional de Paraplejicos, SESCAM, Toledo, Spain
| |
Collapse
|
18
|
Wichmann TO, Kasch H, Dyrskog S, Høy K, Møller BK, Krog J, Hviid CVB, Hoffmann HJ, Rasmussen MM. The inflammatory response and blood-spinal cord barrier integrity in traumatic spinal cord injury: a prospective pilot study. Acta Neurochir (Wien) 2022; 164:3143-3153. [PMID: 36190569 DOI: 10.1007/s00701-022-05369-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 09/07/2022] [Indexed: 02/01/2023]
Abstract
PURPOSE Triggering of inflammatory responses and disruption of blood-spinal cord barrier (BSCB) integrity are considered pivotal events in the pathophysiology of traumatic spinal cord injury (TSCI). Yet, these events are poorly understood and described in humans. This study aims to describe inflammatory responses and BSCB integrity in human TSCI. METHODS Fifteen TSCI patients and fifteen non-TSCI patients were prospectively recruited from Aarhus University Hospital, Denmark. Peripheral blood (PB) and cerebrospinal fluid (CSF) were collected at median day 0 [IQR: 1], median day 9 [IQR: 2], and median day 148 [IQR: 49] after injury. PB and CSF were analyzed for immune cells by flow cytometry, cytokines by multiplex immunoassay, and BSCB integrity by IgG Index. RESULTS Eleven TSCI patients completed follow-up. Results showed alterations in innate and adaptive immune cell counts over time. TSCI patients had significantly increased cytokine concentrations in CSF at the first and second follow-up, while only concentrations of interleukin (IL)-4, IL-8, and tumor necrosis factor-α remained significantly increased at the third follow-up. In PB, TSCI patients had significantly increased IL-6, IL-8, and IL-10 concentrations and significantly decreased interferon-γ concentrations at the first follow-up. Results further showed increased IgG Index indicative of BSCB disruption in seven TSCI patients at the first follow-up, five TSCI patients at the second follow-up, and two patients at the third follow-up. CONCLUSIONS Our results suggest that TSCI mainly triggers innate inflammatory responses that resolves over time, although with some degree of non-resolving inflammation, particularly in CSF. Our results cannot confirm BSCB disruption in all TSCI patients.
Collapse
Affiliation(s)
- Thea Overgaard Wichmann
- Dept. Neurosurgery, Cense-Spine, Aarhus University Hospital, Palle Juul-Jensens, Boulevard 165 8200 Aarhus N, Aarhus, Denmark.
| | - Helge Kasch
- Dept. Neurology, Aarhus University Hospital, Aarhus, Denmark.,Dept. of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Stig Dyrskog
- Dept. Intensive Care, Aarhus University Hospital, Aarhus, Denmark
| | - Kristian Høy
- Dept. of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Dept. Orthopaedic Surgery - Spine section, Aarhus University Hospital, Aarhus, Denmark
| | - Bjarne Kuno Møller
- Dept. of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | - Jan Krog
- Dept. Anaesthesiology, Aarhus University Hospital, Aarhus, Denmark
| | - Claus Vinter Bødker Hviid
- Dept. Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark.,Dept. Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
| | - Hans Jürgen Hoffmann
- Dept. of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Dept. Respiratory Diseases and Allergy, Aarhus University Hospital, Aarhus, Denmark
| | - Mikkel Mylius Rasmussen
- Dept. Neurosurgery, Cense-Spine, Aarhus University Hospital, Palle Juul-Jensens, Boulevard 165 8200 Aarhus N, Aarhus, Denmark.,Dept. of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
19
|
Li C, Wu C, Xu G, Liu Y, Chen J, Zhang J, Hong H, Ji C, Cui Z. CCR7-mediated T follicular helper cell differentiation is associated with the pathogenesis and immune microenvironment of spinal cord injury-induced immune deficiency syndrome. Front Neurosci 2022; 16:1019406. [PMCID: PMC9615471 DOI: 10.3389/fnins.2022.1019406] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Spinal cord injury-induced immune deficiency syndrome (SCI-IDS) is a disorder characterized by systemic immunosuppression secondary to SCI that dramatically increases the likelihood of infection and is difficult to treat. T follicular helper (Tfh) cells regulated by chemokine receptor CCR7 are associated with SCI-IDS after acute SCI. The present study explored the roles of CCR7 in SCI-IDS occurrence and immune microenvironment composition. Gene expression profile data of peripheral blood leukocytes from SCI and non-SCI subjects were collected from the Gene Expression Omnibus database. According to differential gene expression analysis, a protein-protein interaction (PPI) network, and risk model construction, the CCR7 expression level was prominently related to acute SCI and CCR7 expression was significantly downregulated after acute SCI. Next, we constructed a clinical prediction model and used it to identify patients with acute SCI. Using Gene Ontology (GO) analysis and gene set enrichment analysis (GSEA), we discovered that immune-related biological processes, such as T cell receptor signaling pathway, were suppressed, whereas chemokine-related signaling pathways were activated after acute SCI. Immune infiltration analysis performed using single sample GSEA and CIBERSORT suggested that Tfh cell function was significantly correlated with the CCR7 expression levels and was considerably reduced after acute SCI. Acute SCI was divided into two subtypes, and we integrated multiple classifiers to analyze and elucidate the immunomodulatory relationships in both subtypes jointly. The results suggested that CCR7 suppresses the immunodeficiency phenotype by activating the chemokine signaling pathway in Tfh cells. In conclusion, CCR7 exhibits potential as a diagnostic marker for acute SCI.
Collapse
Affiliation(s)
- Chaochen Li
- The First People’s Hospital of Nantong, The Second Affiliated Hospital of Nantong University, Nantong University, Nantong, China
- Key Laboratory for Restoration Mechanism and Clinical Translation of Spinal Cord Injury, Nantong, China
- Research Institute for Spine and Spinal Cord Disease of Nantong University, Nantong, China
| | - Chunshuai Wu
- The First People’s Hospital of Nantong, The Second Affiliated Hospital of Nantong University, Nantong University, Nantong, China
- Key Laboratory for Restoration Mechanism and Clinical Translation of Spinal Cord Injury, Nantong, China
- Research Institute for Spine and Spinal Cord Disease of Nantong University, Nantong, China
| | - Guanhua Xu
- The First People’s Hospital of Nantong, The Second Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| | - Yang Liu
- The First People’s Hospital of Nantong, The Second Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| | - Jiajia Chen
- The First People’s Hospital of Nantong, The Second Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| | - Jinlong Zhang
- The First People’s Hospital of Nantong, The Second Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| | - Hongxiang Hong
- The First People’s Hospital of Nantong, The Second Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| | - Chunyan Ji
- The First People’s Hospital of Nantong, The Second Affiliated Hospital of Nantong University, Nantong University, Nantong, China
- Key Laboratory for Restoration Mechanism and Clinical Translation of Spinal Cord Injury, Nantong, China
- Research Institute for Spine and Spinal Cord Disease of Nantong University, Nantong, China
| | - Zhiming Cui
- The First People’s Hospital of Nantong, The Second Affiliated Hospital of Nantong University, Nantong University, Nantong, China
- Key Laboratory for Restoration Mechanism and Clinical Translation of Spinal Cord Injury, Nantong, China
- Research Institute for Spine and Spinal Cord Disease of Nantong University, Nantong, China
- *Correspondence: Zhiming Cui,
| |
Collapse
|
20
|
Acute Systemic White Blood Cell Changes following Degenerative Cervical Myelopathy (DCM) in a Mouse Model. Int J Mol Sci 2022; 23:ijms231911496. [PMID: 36232808 PMCID: PMC9570488 DOI: 10.3390/ijms231911496] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 09/20/2022] [Accepted: 09/23/2022] [Indexed: 11/29/2022] Open
Abstract
Degenerative cervical myelopathy (DCM) is caused by age-related degeneration of the cervical spine, causing chronic spinal cord compression and inflammation. The aim of this study was to assess whether the natural progression of DCM is accompanied by hematological changes in the white blood cell composition. If so, these changes can be used for diagnosis complementing established imaging approaches and for the development of treatment strategies, since peripheral immunity affects the progression of DCM. Gradual compression of the spinal cord was induced in C57B/L mice at the C5-6 level. The composition of circulating white blood cells was analyzed longitudinally at four time points after induction of DCM using flow cytometry. At 12 weeks, serum cytokine levels were measured using a Luminex x-MAP assay. Neurological impairment in the mouse model was also assessed using the ladder walk test and CatWalk. Stepping function (* p < 0.05) and overground locomotion (*** p < 0.001) were impaired in the DCM group. Importantly, circulating monocytes and T cells were affected primarily at 3 weeks following DCM. T cells were two-fold lower in the DCM group (*** p < 0.0006), whereas monocytes were four-fold increased (*** p < 0.0006) in the DCM compared with the sham group. Our data suggest that changes in white blood cell populations are modest, which is unique to other spinal cord pathologies, and precede the development of neurobehavioral symptoms.
Collapse
|
21
|
Freyermuth-Trujillo X, Segura-Uribe JJ, Salgado-Ceballos H, Orozco-Barrios CE, Coyoy-Salgado A. Inflammation: A Target for Treatment in Spinal Cord Injury. Cells 2022; 11:2692. [PMID: 36078099 PMCID: PMC9454769 DOI: 10.3390/cells11172692] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/16/2022] [Accepted: 08/25/2022] [Indexed: 12/15/2022] Open
Abstract
Spinal cord injury (SCI) is a significant cause of disability, and treatment alternatives that generate beneficial outcomes and have no side effects are urgently needed. SCI may be treatable if intervention is initiated promptly. Therefore, several treatment proposals are currently being evaluated. Inflammation is part of a complex physiological response to injury or harmful stimuli induced by mechanical, chemical, or immunological agents. Neuroinflammation is one of the principal secondary changes following SCI and plays a crucial role in modulating the pathological progression of acute and chronic SCI. This review describes the main inflammatory events occurring after SCI and discusses recently proposed potential treatments and therapeutic agents that regulate inflammation after insult in animal models.
Collapse
Affiliation(s)
- Ximena Freyermuth-Trujillo
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City CP 06720, Mexico
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City CP 04510, Mexico
| | - Julia J. Segura-Uribe
- Subdirección de Gestión de la Investigación, Hospital Infantil de México Federico Gómez, Secretaría de Salud, Mexico City CP 06720, Mexico
| | - Hermelinda Salgado-Ceballos
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City CP 06720, Mexico
| | - Carlos E. Orozco-Barrios
- CONACyT-Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City CP 06720, Mexico
| | - Angélica Coyoy-Salgado
- CONACyT-Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City CP 06720, Mexico
| |
Collapse
|
22
|
Hamilton AM, Sampson TR. Traumatic spinal cord injury and the contributions of the post-injury microbiome. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2022; 167:251-290. [PMID: 36427958 DOI: 10.1016/bs.irn.2022.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Spinal cord injuries are an enormous burden on injured individuals and their caregivers. The pathophysiological effects of injury are not limited to the spine and limb function, but affect numerous body systems. Growing observations in human studies and experimental models suggest that the gut microbiome is altered following spinal cord injury. Given the importance of signals derived from the gut microbiome for host physiology, it is possible that injury-triggered dysbiosis subsequently affects aspects of recovery. Here, we review emerging literature on the role of the microbiome following spinal cord injury. Specifically, we highlight findings from both human and experimental studies that correlate taxonomic changes to aspects of injury recovery. Examination of both observational and emerging interventional studies supports the notion that future therapeutic avenues for spinal cord injury pathologies may lie at the interface of the host and indigenous microbes.
Collapse
Affiliation(s)
- Adam M Hamilton
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Timothy R Sampson
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States.
| |
Collapse
|
23
|
Takamiya S, Kawabori M, Yamazaki K, Yamaguchi S, Tanimori A, Yamamoto K, Ohnishi S, Seki T, Konno K, Tha KK, Hashimoto D, Watanabe M, Houkin K, Fujimura M. Intravenous transplantation of amnion-derived mesenchymal stem cells promotes functional recovery and alleviates intestinal dysfunction after spinal cord injury. PLoS One 2022; 17:e0270606. [PMID: 35802703 PMCID: PMC9269969 DOI: 10.1371/journal.pone.0270606] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 06/13/2022] [Indexed: 11/19/2022] Open
Abstract
Spinal cord injury (SCI) is often accompanied by gastrointestinal dysfunction due to the disconnection of the spinal autonomic nervous system. Gastrointestinal dysfunction reportedly upregulates intestinal permeability, leading to bacterial translocation of the gut microbiome to the systemic circulation, which further activates systemic inflammation, exacerbating neuronal damage. Mesenchymal stem cells (MSC) reportedly ameliorate SCI. Here, we aimed to investigate their effect on the associated gastrointestinal dysfunction. Human amnion-derived MSC (AMSCs) were intravenously transplanted one day after a rat model of midthoracic SCI. Biodistribution of transplanted cells, behavioral assessment, and histological evaluations of the spinal cord and intestine were conducted to elucidate the therapeutic effect of AMSCs. Bacterial translocation of the gut microbiome was examined by in situ hybridization and bacterial culture of the liver. Systemic inflammations were examined by blood cytokines, infiltrating immune cells in the spinal cord, and the size of the peripheral immune tissue. AMSCs released various neurotrophic factors and were mainly distributed in the liver and lung after transplantation. AMSC-transplanted animals showed smaller spinal damage and better neurological recovery with preserved neuronal tract. AMSCs transplantation ameliorated intestinal dysfunction both morphologically and functionally, which prevented translocation of the gut microbiome to the systemic circulation. Systemic inflammations were decreased in animals receiving AMSCs in the chronic phase. Intravenous AMSC administration during the acute phase of SCI rescues both spinal damage and intestinal dysfunction. Reducing bacterial translocation may contribute to decreasing systemic inflammation.
Collapse
Affiliation(s)
- Soichiro Takamiya
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Masahito Kawabori
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
- * E-mail:
| | - Kazuyoshi Yamazaki
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Sho Yamaguchi
- Regenerative Medicine and Cell Therapy Laboratories, Kaneka Corporation, Kobe, Hyogo, Japan
| | - Aki Tanimori
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Koji Yamamoto
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Shunsuke Ohnishi
- Laboratory of Molecular and Cellular Medicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Toshitaka Seki
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Kotaro Konno
- Department of Anatomy and Embryology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Khin Khin Tha
- Global Center for Biomedical Science and Engineering, Hokkaido University Faculty of Medicine, Sapporo, Hokkaido, Japan
| | - Daigo Hashimoto
- Department of Hematology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Masahiko Watanabe
- Department of Anatomy and Embryology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Kiyohiro Houkin
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Miki Fujimura
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| |
Collapse
|
24
|
Fraussen J, Beckers L, van Laake-Geelen CCM, Depreitere B, Deckers J, Cornips EMJ, Peuskens D, Somers V. Altered Circulating Immune Cell Distribution in Traumatic Spinal Cord Injury Patients in Relation to Clinical Parameters. Front Immunol 2022; 13:873315. [PMID: 35837411 PMCID: PMC9273975 DOI: 10.3389/fimmu.2022.873315] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 06/01/2022] [Indexed: 11/13/2022] Open
Abstract
Following a spinal cord injury (SCI), an inflammatory immune reaction is triggered which results in advanced secondary tissue damage. The systemic post-SCI immune response is poorly understood. This study aimed to extensively analyse the circulating immune cell composition in traumatic SCI patients in relation to clinical parameters. High-dimensional flow cytometry was performed on peripheral blood mononuclear cells of 18 traumatic SCI patients and 18 healthy controls to determine immune cell subsets. SCI blood samples were collected at multiple time points in the (sub)acute (0 days to 3 weeks post-SCI, (s)aSCI) and chronic (6 to >18 weeks post-SCI, cSCI) disease phase. Total and CD4+ T cell frequencies were increased in cSCI patients. Both CD4+ T cells and B cells were shifted towards memory phenotypes in (s)aSCI patients and cSCI patients, respectively. Most profound changes were observed in the B cell compartment. Decreased immunoglobulin (Ig)G+ and increased IgM+ B cell frequencies reflected disease severity, as these correlated with American Spinal Injury Association (ASIA) impairment scale (AIS) scores. Post-SCI B cell responses consisted of an increased frequency of CD74+ cells and CD74 expression level within total B cells and B cell subsets. Findings from this study suggest that post-SCI inflammation is driven by memory immune cell subsets. The increased CD74 expression on post-SCI B cells could suggest the involvement of CD74-related pathways in neuroinflammation following SCI. In addition, the clinical and prognostic value of monitoring circulating IgM+ and IgG+ B cell levels in SCI patients should be further evaluated.
Collapse
Affiliation(s)
- Judith Fraussen
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - Lien Beckers
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - Charlotte C. M. van Laake-Geelen
- Adelante Centre of Expertise in Rehabilitation and Audiology, Hoensbroek, Netherlands
- Department of Rehabilitation Medicine, Research School CAPHRI, Maastricht University, Maastricht, Netherlands
| | - Bart Depreitere
- Division of Neurosurgery, University Hospitals Leuven, Leuven, Belgium
| | - Jens Deckers
- Department of Neurosurgery, Algemeen Ziekenhuis (AZ) Turnhout, Turnhout, Belgium
- Department of Neurosurgery, Ziekenhuis Oost-Limburg, Genk, Belgium
| | | | - Dieter Peuskens
- Department of Neurosurgery, Ziekenhuis Oost-Limburg, Genk, Belgium
| | - Veerle Somers
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- *Correspondence: Veerle Somers,
| |
Collapse
|
25
|
Bloom O, Tracey KJ, Pavlov VA. Exploring the vagus nerve and the inflammatory reflex for therapeutic benefit in chronic spinal cord injury. Curr Opin Neurol 2022; 35:249-257. [PMID: 35102123 PMCID: PMC9258775 DOI: 10.1097/wco.0000000000001036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW To describe features and implications of chronic systemic inflammation in individuals with spinal cord injury (SCI) and to summarize the growing therapeutic possibilities to explore the vagus nerve-mediated inflammatory reflex in this context. RECENT FINDINGS The discovery of the inflammatory reflex provides a rationale to explore neuromodulation modalities, that is, electrical vagus nerve stimulation and pharmacological cholinergic modalities to regulate inflammation after SCI. SUMMARY Inflammation in individuals with SCI may negatively impact functional recovery and medical consequences after SCI. Exploring the potential of the vagus nerve-based inflammatory reflex to restore autonomic regulation and control inflammation may provide a novel approach for functional improvement in SCI.
Collapse
Affiliation(s)
- Ona Bloom
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset
- Donald and Barbara Zucker School of Medicine, Hempstead, New York, USA
| | - Kevin J. Tracey
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset
- Donald and Barbara Zucker School of Medicine, Hempstead, New York, USA
| | - Valentin A. Pavlov
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset
- Donald and Barbara Zucker School of Medicine, Hempstead, New York, USA
| |
Collapse
|
26
|
Garcia E, Hernández-Ayvar F, Rodríguez-Barrera R, Flores-Romero A, Borlongan C, Ibarra A. Supplementation With Vitamin E, Zinc, Selenium, and Copper Re-Establishes T-Cell Function and Improves Motor Recovery in a Rat Model of Spinal Cord Injury. Cell Transplant 2022; 31:9636897221109884. [PMID: 35808825 PMCID: PMC9272473 DOI: 10.1177/09636897221109884] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Spinal cord injury (SCI) causes a dysfunction of sympathetic nervous
system innervation that affects the immune system, leading to
immunosuppression syndrome (ISS) and contributing to patient
degeneration and increased risk of several infections. A possible
therapeutic strategy that could avoid further patient deterioration is
the supplementation with Vitamin E or trace elements, such as Zinc,
Selenium, and Copper, which individually promotes T-cell
differentiation and proliferative responses. For this reason, the aim
of the present study was to evaluate whether Vitamin E, Zinc,
Selenium, and Copper supplementation preserves the number of
T-lymphocytes and improves their proliferative function after
traumatic SCI. Sprague–Dawley female rats were subjected to moderate
SCI and then randomly allocated into three groups: (1) SCI +
supplements; (2) SCI + vehicle (olive oil and phosphate-buffered
saline); and (3) sham-operated rats. In all rats, the intervention was
initiated 15 min after SCI and then administered daily until the end
of study. Locomotor recovery was assessed at 7 and 15 days after SCI.
At 15 days after supplementation, the quantification of the number of
T-cells and its proliferation function were examined. Our results
showed that the SCI + supplements group presented a significant
improvement in motor recovery at 7 and 15 days after SCI. In addition,
this group showed a better T-cell number and proliferation rate than
that observed in the group with SCI + vehicle. Our findings suggest
that Vitamin E, Zinc, Selenium, and Copper supplementation could be
part of a therapy for patients suffering from acute SCI, helping to
preserve T-cell function, avoiding complications, and promoting a
better motor recovery. All procedures were approved by the Animal
Bioethics and Welfare Committee (Approval No. 201870; CSNBTBIBAJ
090812960).
Collapse
Affiliation(s)
- Elisa Garcia
- Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucan, México
| | - Fernanda Hernández-Ayvar
- Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucan, México
| | - Roxana Rodríguez-Barrera
- Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucan, México
| | - Adrián Flores-Romero
- Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucan, México
| | - Cesar Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Antonio Ibarra
- Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucan, México
| |
Collapse
|
27
|
Krebs J, Stoyanov J, Wöllner J, Valido E, Pannek J. Immunomodulation for primary prevention of urinary tract infections in patients with spinal cord injury during primary rehabilitation: protocol for a randomized placebo-controlled pilot trial (UROVAXOM-pilot). Trials 2021; 22:677. [PMID: 34607600 PMCID: PMC8489057 DOI: 10.1186/s13063-021-05630-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 09/15/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Urinary tract infections (UTIs) are common in individuals with neurogenic lower urinary tract dysfunction (NLUTD) following spinal cord injury (SCI). They are not only a great burden for affected individuals, but also cause considerable health costs. Furthermore, recurrent antibiotic treatments of UTIs contribute to the growing problem of bacterial resistance to antimicrobial compounds. Even though there is a multitude of different measures to prevent UTIs in individuals with NLUTD, no clear evidence exists for any of these. Oral immunomodulation with UTI-relevant Escherichia coli lysate may be a promising preventative measure with a good safety profile in individuals with NLUTD. However, currently available data are sparse. METHODS This is a randomized, quasi-blinded, placebo-controlled, mono-centric pilot trial investigating the feasibility of a main trial regarding the effects of a lyophilized lysate of E. coli strains for oral application (Uro-Vaxom®, OM Pharma SA, Meyrin, Switzerland). There will be two parallel groups of 12 participants each. Individuals with acute SCI (duration SCI ≤ 56 days) from 18 to 70 years of age admitted for primary rehabilitation will be eligible. Blood and urine samples will be taken prior to intervention start, at the end of the intervention, and 3 months after intervention termination. The trial intervention will last 90 days. The participants will not be informed regarding the treatment allocation (quasi-blinded). The nursing staff will prepare the daily dose of the allocated treatment from the original packaging. The trial personnel and the biostatistician will be blinded. Feasibility (e.g., recruitment rate, patient attrition), clinical (e.g., number of symptomatic UTIs), and laboratory parameters (e.g., urinary culture, urinary proteo- and microbiome, blood cell counts) as well as adverse events will be collected. DISCUSSION Effective and efficient measures for the prevention of UTIs in individuals with NLUTD are urgently needed. If the conclusion of this pilot is positive regarding feasibility, the effects of oral immunomodulation with a E. coli lysate will be investigated in a larger, sufficiently powered, multi-center trial. TRIAL REGISTRATION ClinicalTrials.gov NCT04049994 . Registered on 8 August 2019.
Collapse
Affiliation(s)
- Jörg Krebs
- Clinical Trial Unit, Swiss Paraplegic Centre, Nottwil, Switzerland.
| | - Jivko Stoyanov
- SCI Population Biobanking & Translational Medicine Group, Swiss Paraplegic Research, Nottwil, Switzerland
- Institute of Social and Preventive Medicine (ISPM), University of Bern, Bern, Switzerland
| | - Jens Wöllner
- Neuro-Urology, Swiss Paraplegic Centre, Nottwil, Switzerland
| | - Ezra Valido
- SCI Population Biobanking & Translational Medicine Group, Swiss Paraplegic Research, Nottwil, Switzerland
| | - Jürgen Pannek
- Neuro-Urology, Swiss Paraplegic Centre, Nottwil, Switzerland
- Department of Urology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
28
|
Debaud C, Tseng HW, Chedik M, Kulina I, Genêt F, Ruitenberg MJ, Levesque JP. Local and Systemic Factors Drive Ectopic Osteogenesis in Regenerating Muscles of Spinal-Cord-Injured Mice in a Lesion-Level-Dependent Manner. J Neurotrauma 2021; 38:2162-2175. [PMID: 33913747 DOI: 10.1089/neu.2021.0058] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Neuroimmune dysfunction is thought to promote the development of several acute and chronic complications in spinal cord injury (SCI) patients. Putative roles for adrenal stress hormones and catecholamines are increasingly being recognized, yet how these adversely affect peripheral tissue homeostasis and repair under SCI conditions remains elusive. Here, we investigated their influence in a mouse model of SCI with acquired neurogenic heterotopic ossification. We show that spinal cord lesions differentially influence muscular regeneration in a level-dependent manner and through a complex multi-step process that creates an osteopermissive environment within the first hours of injury. This cascade of events is shown to critically involve adrenergic signals and drive the acute release of the neuropeptide, substance P. Our findings generate new insights into the kinetics and processes that govern SCI-induced deregulations in skeletal muscle homeostasis and regeneration, thereby aiding the development of sequential therapeutic strategies that can prevent or attenuate neuromusculoskeletal complications in SCI patients.
Collapse
Affiliation(s)
- Charlotte Debaud
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
- Spine Division, Orthopaedic Surgery Department, Queensland Health, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
- Université de Versailles Saint Quentin en Yvelines, U1179 INSERM, UFR des Sciences de la Santé-Simone Veil, Montigny-le-Bretonneux, France
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Saint Lucia, Queensland, Australia
| | - Hsu-Wen Tseng
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Malha Chedik
- Université de Versailles Saint Quentin en Yvelines, U1179 INSERM, UFR des Sciences de la Santé-Simone Veil, Montigny-le-Bretonneux, France
| | - Irina Kulina
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - François Genêt
- Université de Versailles Saint Quentin en Yvelines, U1179 INSERM, UFR des Sciences de la Santé-Simone Veil, Montigny-le-Bretonneux, France
- Service de Réhabilitation, Hôpital Raymond Poincaré, APHP, CIC-IT 1429, Garches, France
| | - Marc J Ruitenberg
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Saint Lucia, Queensland, Australia
| | - Jean-Pierre Levesque
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| |
Collapse
|
29
|
Treelet transform analysis to identify clusters of systemic inflammatory variance in a population with moderate-to-severe traumatic brain injury. Brain Behav Immun 2021; 95:45-60. [PMID: 33524553 PMCID: PMC9004489 DOI: 10.1016/j.bbi.2021.01.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/20/2020] [Accepted: 01/21/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Inflammatory cascades following traumatic brain injury (TBI) can have both beneficial and detrimental effects on recovery. Single biomarker studies do not adequately reflect the major arms of immunity and their relationships to long-term outcomes. Thus, we applied treelet transform (TT) analysis to identify clusters of interrelated inflammatory markers reflecting major components of systemic immune function for which substantial variation exists among individuals with moderate-to-severe TBI. METHODS Serial blood samples from 221 adults with moderate-to-severe TBI were collected over 1-6 months post-injury (n = 607 samples). Samples were assayed for 33 inflammatory markers using Millipore multiplex technology. TT was applied to standardized mean biomarker values generated to identify latent patterns of correlated markers. Treelet clusters (TC) were characterized by biomarkers related to adaptive immunity (TC1), innate immunity (TC2), soluble molecules (TC3), allergy immunity (TC4), and chemokines (TC5). For each TC, a score was generated as the linear combination of standardized biomarker concentrations and cluster load for each individual in the cohort. Ordinal logistic or linear regression was used to test associations between TC scores and 6- and 12-month Glasgow Outcome Scale (GOS), Disability Rating Scale (DRS), and covariates. RESULTS When adjusting for clinical covariates, TC5 was significantly associated with 6-month GOS (odds ratio, OR = 1.44; p-value, p = 0.025) and 6-month DRS scores (OR = 1.46; p = 0.013). TC5 relationships were attenuated when including all TC scores in the model (GOS: OR = 1.29, p = 0.163; DRS: OR = 1.33, p = 0.100). When adjusting for all TC scores and covariates, only TC3 was associated with 6- and 12-month GOS (OR = 1.32, p = 0.041; OR = 1.39, p = 0.002) and also 6- and 12-month DRS (OR = 1.38, p = 0.016; OR = 1.58, p = 0.0002). When applying TT to inflammation markers significantly associated with 6-month GOS, multivariate modeling confirmed that TC3 remained significantly associated with GOS. Biomarker cluster membership remained consistent between the GOS-specific dendrogram and overall dendrogram. CONCLUSIONS TT effectively characterized chronic, systemic immunity among a cohort of individuals with moderate-to-severe TBI. We posit that chronic chemokine levels are effector molecules propagating cellular immune dysfunction, while chronic soluble receptors are inflammatory damage readouts perpetuated, in part, by persistent dysfunctional cellular immunity to impact neuro-recovery.
Collapse
|
30
|
Beyond the lesion site: minocycline augments inflammation and anxiety-like behavior following SCI in rats through action on the gut microbiota. J Neuroinflammation 2021; 18:144. [PMID: 34174901 PMCID: PMC8234629 DOI: 10.1186/s12974-021-02123-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 03/03/2021] [Indexed: 12/17/2022] Open
Abstract
Background Minocycline is a clinically available synthetic tetracycline derivative with anti-inflammatory and antibiotic properties. The majority of studies show that minocycline can reduce tissue damage and improve functional recovery following central nervous system injuries, mainly attributed to the drug’s direct anti-inflammatory, anti-oxidative, and neuroprotective properties. Surprisingly the consequences of minocycline’s antibiotic (i.e., antibacterial) effects on the gut microbiota and systemic immune response after spinal cord injury have largely been ignored despite their links to changes in mental health and immune suppression. Methods Here, we sought to determine minocycline’s effect on spinal cord injury-induced changes in the microbiota-immune axis using a cervical contusion injury in female Lewis rats. We investigated a group that received minocycline following spinal cord injury (immediately after injury for 7 days), an untreated spinal cord injury group, an untreated uninjured group, and an uninjured group that received minocycline. Plasma levels of cytokines/chemokines and fecal microbiota composition (using 16s rRNA sequencing) were monitored for 4 weeks following spinal cord injury as measures of the microbiota-immune axis. Additionally, motor recovery and anxiety-like behavior were assessed throughout the study, and microglial activation was analyzed immediately rostral to, caudal to, and at the lesion epicenter. Results We found that minocycline had a profound acute effect on the microbiota diversity and composition, which was paralleled by the subsequent normalization of spinal cord injury-induced suppression of cytokines/chemokines. Importantly, gut dysbiosis following spinal cord injury has been linked to the development of anxiety-like behavior, which was also decreased by minocycline. Furthermore, although minocycline attenuated spinal cord injury-induced microglial activation, it did not affect the lesion size or promote measurable motor recovery. Conclusion We show that minocycline’s microbiota effects precede its long-term effects on systemic cytokines and chemokines following spinal cord injury. These results provide an exciting new target of minocycline as a therapeutic for central nervous system diseases and injuries. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02123-0.
Collapse
|
31
|
Ueno M. Restoring neuro-immune circuitry after brain and spinal cord injuries. Int Immunol 2021; 33:311-325. [PMID: 33851981 DOI: 10.1093/intimm/dxab017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 04/13/2021] [Indexed: 12/17/2022] Open
Abstract
Neuro-immune interactions are essential for our body's defense and homeostasis. Anatomical and physiological analyses have shown that the nervous system comprises multiple pathways that regulate the dynamics and functions of immune cells, which are mainly mediated by the autonomic nervous system and adrenal signals. These are disturbed when the neurons and circuits are damaged by diseases of the central nervous system (CNS). Injuries caused by stroke or trauma often cause immune dysfunction by abrogation of the immune-regulating neural pathways, which leads to an increased risk of infections. Here, I review the structures and functions of the neural pathways connecting the brain and the immune system, and the neurogenic mechanisms of immune dysfunction that emerge after CNS injuries. Recent technological advances in manipulating specific neural circuits have added mechanistic aspects of neuro-immune interactions and their dysfunctions. Understanding the neural bases of immune control and their pathological processes will deepen our knowledge of homeostasis and lead to the development of strategies to cure immune deficiencies observed in various CNS disorders.
Collapse
Affiliation(s)
- Masaki Ueno
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, Niigata 951-8585, Japan
| |
Collapse
|
32
|
Aschauer-Wallner S, Leis S, Bogdahn U, Johannesen S, Couillard-Despres S, Aigner L. Granulocyte colony-stimulating factor in traumatic spinal cord injury. Drug Discov Today 2021; 26:1642-1655. [PMID: 33781952 DOI: 10.1016/j.drudis.2021.03.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 02/23/2021] [Accepted: 03/17/2021] [Indexed: 12/12/2022]
Abstract
Granulocyte colony-stimulating factor (G-CSF) is a cytokine used in pharmaceutical preparations for the treatment of chemotherapy-induced neutropenia. Evidence from experimental studies indicates that G-CSF exerts relevant activities in the central nervous system (CNS) in particular after lesions. In acute, subacute, and chronic CNS lesions, G-CSF appears to have strong anti-inflammatory, antiapoptotic, antioxidative, myelin-protective, and axon-regenerative activities. Additional effects result in the stimulation of angiogenesis and neurogenesis as well as in bone marrow stem cell mobilization to the CNS. There are emerging preclinical and clinical data indicating that G-CSF is a safe and effective drug for the treatment of acute and chronic traumatic spinal cord injury (tSCI), which we summarize in this review.
Collapse
Affiliation(s)
- Stephanie Aschauer-Wallner
- Department of Orthopedics and Traumatology, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria; Institute of Molecular Regenerative Medicine, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria.
| | - Stefan Leis
- Department of Neurology, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Ulrich Bogdahn
- Velvio GmbH, Regensburg, Germany; Department of Neurology, University Hospital Regensburg, Regensburg, Germany
| | - Siw Johannesen
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany; Department of Neurology, BG Trauma Center Murnau, Murnau, Germany
| | - Sebastien Couillard-Despres
- Institute of Experimental Neuroregeneration, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
33
|
Chio JCT, Xu KJ, Popovich P, David S, Fehlings MG. Neuroimmunological therapies for treating spinal cord injury: Evidence and future perspectives. Exp Neurol 2021; 341:113704. [PMID: 33745920 DOI: 10.1016/j.expneurol.2021.113704] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/01/2021] [Accepted: 03/16/2021] [Indexed: 12/13/2022]
Abstract
Spinal cord injury (SCI) has a complex pathophysiology. Following the initial physical trauma to the spinal cord, which may cause vascular disruption, hemorrhage, mechanical injury to neural structures and necrosis, a series of biomolecular cascades is triggered to evoke secondary injury. Neuroinflammation plays a major role in the secondary injury after traumatic SCI. To date, the administration of systemic immunosuppressive medications, in particular methylprednisolone sodium succinate, has been the primary pharmacological treatment. This medication is given as a complement to surgical decompression of the spinal cord and maintenance of spinal cord perfusion through hemodynamic augmentation. However, the impact of neuroinflammation is complex with harmful and beneficial effects. The use of systemic immunosuppressants is further complicated by the natural onset of post-injury immunosuppression, which many patients with SCI develop. It has been hypothesized that immunomodulation to attenuate detrimental aspects of neuroinflammation after SCI, while avoiding systemic immunosuppression, may be a superior approach. To accomplish this, a detailed understanding of neuroinflammation and the systemic immune responses after SCI is required. Our review will strive to achieve this goal by first giving an overview of SCI from a clinical and basic science context. The role that neuroinflammation plays in the pathophysiology of SCI will be discussed. Next, the positive and negative attributes of the innate and adaptive immune systems in neuroinflammation after SCI will be described. With this background established, the currently existing immunosuppressive and immunomodulatory therapies for treating SCI will be explored. We will conclude with a summary of topics that can be explored by neuroimmunology research. These concepts will be complemented by points to be considered by neuroscientists developing therapies for SCI and other injuries to the central nervous system.
Collapse
Affiliation(s)
- Jonathon Chon Teng Chio
- Division of Translational and Experimental Neuroscience, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.
| | - Katherine Jiaxi Xu
- Human Biology Program, University of Toronto, Wetmore Hall, 300 Huron St., Room 105, Toronto, Ontario M5S 3J6, Canada.
| | - Phillip Popovich
- Department of Neuroscience, Belford Center for Spinal Cord Injury, Center for Brain and Spinal Cord Repair, The Neurological Institute, The Ohio State University, Wexner Medical Center, 410 W. 10(th) Ave., Columbus 43210, USA.
| | - Samuel David
- Centre for Research in Neuroscience and BRaIN Program, The Research Institute of the McGill University Health Centre, 1650 Cedar Ave., Montreal, Quebec H3G 1A4, Canada.
| | - Michael G Fehlings
- Division of Translational and Experimental Neuroscience, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
34
|
Sahbani K, Shultz LC, Cardozo CP, Bauman WA, Tawfeek HA. Absence of αβ T cells accelerates disuse bone loss in male mice after spinal cord injury. Ann N Y Acad Sci 2021; 1487:43-55. [PMID: 33107070 DOI: 10.1111/nyas.14518] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/07/2020] [Accepted: 10/07/2020] [Indexed: 11/30/2022]
Abstract
Whether T cells promote bone loss following immobilization after spinal cord injury (SCI) remains undetermined. Therefore, wild-type (WT) and T cell-deficient (Tcrb-/- ) male mice underwent sham or contusion SCI to cause hindlimb paralysis. Femurs were isolated and distal and midshaft regions were evaluated by microcomputed tomography scanning. Bone marrow (BM) levels of bone turnover markers, as well as receptor activator of nuclear factor-kappa B ligand (RANKL) and osteoprotegerin (OPG), were measured by ELISA. At 2 weeks post-SCI, immobilization resulted in marked reduction in trabecular fractional bone volume (55%), thickness (40%), connectivity, and cortical thickness only in the Tcrb-/- animals (interaction with P < 0.05). BM analysis revealed lower bone formation (procollagen type 1 intact N-terminal propeptide), higher bone resorption (tartrate-resistant acid phosphatase-5b), and a higher RANKL/OPG ratio in the Tcrb-/- SCI animals. At 5 weeks post-SCI, while both WT and Tcrb-/- paralyzed animals showed deterioration of all indices of bone structure, they were more severe in Tcrb-/- animals. In summary, unlike other skeletal disorders, loss of αβ T cells compromises, rather than preserves, skeletal integrity under conditions of immobilization.
Collapse
MESH Headings
- Animals
- Bone Density/genetics
- Bone Density/immunology
- Bone Diseases, Metabolic/genetics
- Bone Diseases, Metabolic/immunology
- Bone Diseases, Metabolic/metabolism
- Bone Diseases, Metabolic/pathology
- Bone Resorption/genetics
- Bone Resorption/immunology
- Bone Resorption/metabolism
- Cell Count
- Genes, T-Cell Receptor beta/genetics
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Receptors, Antigen, T-Cell, alpha-beta/deficiency
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Spinal Cord Injuries/complications
- Spinal Cord Injuries/genetics
- Spinal Cord Injuries/immunology
- Spinal Cord Injuries/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/pathology
- X-Ray Microtomography
Collapse
Affiliation(s)
- Karim Sahbani
- National Center for the Medical Consequences of Spinal Cord Injury, James J Peters Veterans Affairs Medical Center, Bronx, New York
- Bronx Veterans Medical Research Foundation Inc., Bronx, New York
| | - Laura C Shultz
- Veterinary Medical Unit, James J Peters Veterans Affairs Medical Center, Bronx, New York
| | - Christopher P Cardozo
- National Center for the Medical Consequences of Spinal Cord Injury, James J Peters Veterans Affairs Medical Center, Bronx, New York
- Bronx Veterans Medical Research Foundation Inc., Bronx, New York
- Department of Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Rehabilitation Medicine and Human Performance, The Icahn School of Medicine at Mount Sinai, New York, New York
| | - William A Bauman
- National Center for the Medical Consequences of Spinal Cord Injury, James J Peters Veterans Affairs Medical Center, Bronx, New York
- Bronx Veterans Medical Research Foundation Inc., Bronx, New York
- Department of Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York
| | - Hesham A Tawfeek
- National Center for the Medical Consequences of Spinal Cord Injury, James J Peters Veterans Affairs Medical Center, Bronx, New York
- Bronx Veterans Medical Research Foundation Inc., Bronx, New York
- Department of Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
35
|
Bannerman CA, Douchant K, Sheth PM, Ghasemlou N. The gut-brain axis and beyond: Microbiome control of spinal cord injury pain in humans and rodents. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2021; 9:100059. [PMID: 33426367 PMCID: PMC7779861 DOI: 10.1016/j.ynpai.2020.100059] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/26/2020] [Accepted: 12/10/2020] [Indexed: 12/17/2022]
Abstract
Spinal cord injury (SCI) is a devastating injury to the central nervous system in which 60 to 80% of patients experience chronic pain. Unfortunately, this pain is notoriously difficult to treat, with few effective options currently available. Patients are also commonly faced with various compounding injuries and medical challenges, often requiring frequent hospitalization and antibiotic treatment. Change in the gut microbiome from the "normal" state to one of imbalance, referred to as gut dysbiosis, has been found in both patients and rodent models following SCI. Similarities exist in the bacterial changes observed after SCI and other diseases with chronic pain as an outcome. These changes cause a shift in the regulation of inflammation, causing immune cell activation and secretion of inflammatory mediators that likely contribute to the generation/maintenance of SCI pain. Therefore, correcting gut dysbiosis may be used as a tool towards providing patients with effective pain management and improved quality of life.
Collapse
Affiliation(s)
- Courtney A. Bannerman
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Ontario, Canada
| | - Katya Douchant
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Ontario, Canada
- Gastrointestinal Disease Research Unit, Kingston Health Sciences Center, Kingston, Ontario, Canada
| | - Prameet M. Sheth
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Ontario, Canada
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, Ontario, Canada
- Division of Microbiology, Kingston Health Sciences Centre, Kingston, Ontario, Canada
- Gastrointestinal Disease Research Unit, Kingston Health Sciences Center, Kingston, Ontario, Canada
| | - Nader Ghasemlou
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Ontario, Canada
- Department of Anesthesiology and Perioperative Medicine, Kingston Health Sciences Centre, Kingston, Ontario, Canada
- Centre for Neuroscience Studies, Queen’s University, Kingston, Ontario, Canada
| |
Collapse
|
36
|
Henzel MK, Shultz JM, Dyson‐Hudson TA, Svircev JN, DiMarco AF, Gater DR. Initial assessment and management of respiratory infections in persons with spinal cord injuries and disorders in the COVID-19 era. J Am Coll Emerg Physicians Open 2020; 1:1404-1412. [PMID: 33392545 PMCID: PMC7771758 DOI: 10.1002/emp2.12282] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 09/24/2020] [Accepted: 09/24/2020] [Indexed: 12/13/2022] Open
Abstract
As the COVID-19 pandemic unfolds, emergency department (ED) personnel will face a higher caseload, including those with special medical needs such as persons living with spinal cord injuries and disorders (SCI/D). Individuals with SCI/D who develop COVID-19 are at higher risk for rapid decompensation and development of acute respiratory failure during respiratory infections due to the combination of chronic respiratory muscle paralysis and autonomic dysregulation causing neurogenic restrictive/obstructive lung disease and chronic immune dysfunction. Often, acute respiratory infections will lead to significant mucus production in individuals with SCI/D, and aggressive secretion management is an important component of successful medical treatment. Secretion management techniques include nebulized bronchodilators, chest percussion/drainage techniques, manually assisted coughing techniques, nasotracheal suctioning, and mechanical insufflation-exsufflation. ED professionals, including respiratory therapists, should be familiar with the significant comorbidities associated with SCI/D and the customized secretion management procedures and techniques required for optimal medical management and prevention of respiratory failure. Importantly, protocols should also be implemented to minimize potential COVID-19 spread during aerosol-generating procedures.
Collapse
Affiliation(s)
- M. Kristi Henzel
- Spinal Cord Injury and Disorders ServiceLouis Stokes Cleveland Department of Veterans Affairs Medical CenterClevelandOhioUSA
- Department of Physical Medicine and RehabilitationCase Western Reserve University School of MedicineClevelandOhioUSA
| | - James M. Shultz
- Center for Disaster & Extreme Event Preparedness (DEEP Center)Department of Public Health SciencesUniversity of Miami Leonard M. Miller School of MedicineMiamiFloridaUSA
| | - Trevor A. Dyson‐Hudson
- Center for Spinal Cord Injury ResearchKessler FoundationWest OrangeNew JerseyUSA
- Northern New Jersey Spinal Cord Injury SystemKessler FoundationWest OrangeNew JerseyUSA
- Department of Physical Medicine and RehabilitationRutgers New Jersey Medical SchoolWest OrangeNew JerseyUSA
| | - Jelena N. Svircev
- Department of Veterans Affairs Puget Sound Health Care SystemSpinal Cord Injury ServiceSeattleWashingtonUSA
- Department of Rehabilitation MedicineUniversity of Washington School of MedicineSeattleWashingtonUSA
| | - Anthony F. DiMarco
- Department of Physical Medicine & RehabilitationCase Western Reserve UniversityClevelandOhioUSA
- MetroHealth Medical CenterPulmonary, Sleep Medicine and Critical Care MedicineClevelandOhioUSA
| | - David R. Gater
- Department of Physical Medicine & RehabilitationUniversity of Miami Leonard M. Miller School of MedicineMiamiFloridaUSA
- Christine E. Lynn Rehabilitation CenterMiami Project to Cure Paralysis and Jackson Health SystemMiamiFloridaUSA
| |
Collapse
|
37
|
Chio JCT, Wang J, Surendran V, Li L, Zavvarian MM, Pieczonka K, Fehlings MG. Delayed administration of high dose human immunoglobulin G enhances recovery after traumatic cervical spinal cord injury by modulation of neuroinflammation and protection of the blood spinal cord barrier. Neurobiol Dis 2020; 148:105187. [PMID: 33249350 DOI: 10.1016/j.nbd.2020.105187] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 11/16/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND/INTRODUCTION The neuroinflammatory response plays a major role in the secondary injury cascade after traumatic spinal cord injury (SCI). To date, systemic anti-inflammatory medications such as methylprednisolone sodium succinate (MPSS) have shown promise in SCI. However, systemic immunosuppression can have detrimental side effects. Therefore, immunomodulatory approaches including the use of human immunoglobulin G (hIgG) could represent an attractive alternative. While emerging preclinical data suggests that hIgG is neuroprotective after SCI, the optimal time window of administration and the mechanism of action remain incompletely understood. These knowledge gaps were the focus of this research study. METHODS Female adult Wistar rats received a clip compression-contusion SCI at the C7/T1 level of the spinal cord. Injured rats were randomized, in a blinded manner, to receive a single intravenous bolus of hIgG (2 g/kg) or control buffer at 15 minutes (min), 1 hour (h) or 4 h post-SCI. At 24 h and 8 weeks post-SCI, molecular, histological and neurobehavioral analyses were undertaken. RESULTS At all 3 administration time points, hIgG (2 g/kg) resulted in significantly better short-term and long-term outcomes as compared to control buffer. No significant differences were observed when comparing outcomes between the different time points of administration. At 24 h post-injury, hIgG (2 g/kg) administration enhanced the integrity of the blood spinal cord barrier (BSCB) by increasing expression of tight junction proteins and reducing inflammatory enzyme expression. Improvements in BSCB integrity were associated with reduced immune cell infiltration, lower amounts of albumin and Evans Blue in the injured spinal cord and greater expression of anti-inflammatory cytokines. Furthermore, hIgG (2 g/kg) increased expression of neutrophil chemoattractants in the spleen and sera. After hIgG (2 g/kg) treatment, there were more neutrophils in the spleen and fewer neutrophils in the blood. hIgG also co-localized with endothelial cell ligands that mediate neutrophil extravasation into the injured spinal cord. Importantly, short-term effects of delayed hIgG (2 g/kg) administration were associated with enhanced tissue and neuron preservation, as well as neurobehavioral and sensory recovery at 8 weeks post-SCI. DISCUSSION AND CONCLUSION hIgG (2 g/kg) shows promise as a therapeutic approach for SCI. The anti-inflammatory effects mediated by hIgG (2 g/kg) in the injured spinal cord might be explained in twofold. First, hIgG might antagonize neutrophil infiltration into the spinal cord by co-localizing with endothelial cell ligands that mediate various steps in neutrophil extravasation. Second, hIgG could traffic neutrophils towards the spleen by increasing expression of neutrophil chemoattractants in the spleen and sera. Overall, we demonstrate that delayed administration of hIgG (2 g/kg) at 1 and 4-h post-injury enhances short-term and long-term benefits after SCI by modulating local and systemic neuroinflammatory cascades.
Collapse
Affiliation(s)
- Jonathon Chon Teng Chio
- Division of Translational and Experimental Neuroscience, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.
| | - Jian Wang
- Division of Translational and Experimental Neuroscience, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.
| | - Vithushan Surendran
- Division of Translational and Experimental Neuroscience, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.
| | - Lijun Li
- Division of Translational and Experimental Neuroscience, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.
| | - Mohammad-Masoud Zavvarian
- Division of Translational and Experimental Neuroscience, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.
| | - Katarzyna Pieczonka
- Division of Translational and Experimental Neuroscience, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.
| | - Michael G Fehlings
- Division of Translational and Experimental Neuroscience, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
38
|
Gillespie ER, Ruitenberg MJ. Neuroinflammation after SCI: Current Insights and Therapeutic Potential of Intravenous Immunoglobulin. J Neurotrauma 2020; 39:320-332. [PMID: 32689880 DOI: 10.1089/neu.2019.6952] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Traumatic spinal cord injury (SCI) elicits a complex cascade of cellular and molecular inflammatory events. Although certain aspects of the inflammatory response are essential to wound healing and repair, post-SCI inflammation is, on balance, thought to be detrimental to recovery by causing "bystander damage" and the spread of pathology into spared but vulnerable regions of the spinal cord. Much of the research to date has therefore focused on understanding the inflammatory drivers of secondary tissue loss after SCI, to define therapeutic targets and positively modulate this response. Numerous experimental studies have demonstrated that modulation of the inflammatory response to SCI can indeed lead to significant neuroprotection and improved recovery. However, it is now also recognized that broadscale immunosuppression is not necessarily beneficial and may even carry the risk of contributing to the development of serious adverse events. Immune modulation rather than suppression is therefore now considered a more promising approach to target harmful post-traumatic inflammation following a major neurotraumatic event such as SCI. One promising immunomodulatory agent is intravenous immunoglobulin (IVIG), a plasma product that contains mostly immunoglobulin G (IgG) from thousands of healthy donors. IVIG is currently already widely used to treat a range of autoimmune diseases, but recent studies have found that it also holds great promise for treating acute neurological conditions, including SCI. This review provides an overview of the inflammatory response to SCI, immunomodulatory approaches that are currently in clinical trials, proposed mechanisms of action for IVIG therapy, and the putative relevance of these in the context of neurotraumatic events.
Collapse
Affiliation(s)
- Ellen R Gillespie
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Marc J Ruitenberg
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Australia.,Trauma, Critical Care, and Recovery, Brisbane Diamantina Health Partners, Brisbane, Australia
| |
Collapse
|
39
|
Druschel C, Ossami Saidy RR, Grittner U, Nowak CP, Meisel A, Schaser KD, Niedeggen A, Liebscher T, Kopp MA, Schwab JM. Clinical decision-making on spinal cord injury-associated pneumonia: a nationwide survey in Germany. Spinal Cord 2020; 58:873-881. [PMID: 32071433 PMCID: PMC7223654 DOI: 10.1038/s41393-020-0435-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 01/29/2020] [Accepted: 02/03/2020] [Indexed: 11/13/2022]
Abstract
STUDY DESIGN Survey study. OBJECTIVES Spinal cord injury (SCI)-associated pneumonia (SCI-AP) is associated with poor functional recovery and a major cause of death after SCI. Better tackling SCI-AP requires a common understanding on how SCI-AP is defined. This survey examines clinical algorithms relevant for diagnosis and treatment of SCI-AP. SETTING All departments for SCI-care in Germany. METHODS The clinical decision-making on SCI-AP and the utility of the Centers for Disease Control and Prevention (CDC) criteria for diagnosis of 'clinically defined pneumonia' were assessed by means of a standardized questionnaire including eight case vignettes of suspected SCI-AP. The diagnostic decisions based on the case information were analysed using classification and regression trees (CART). RESULTS The majority of responding departments were aware of the CDC-criteria (88%). In the clinical vignettes, 38-81% of the departments diagnosed SCI-AP in accordance with the CDC-criteria and 7-41% diagnosed SCI-AP in deviation from the CDC-criteria. The diagnostic agreement was not associated with the availability of standard operating procedures for SCI-AP management in the departments. CART analysis identified radiological findings, fever, and worsened gas exchange as most important for the decision on SCI-AP. Frequently requested supplementary diagnostics were microbiological analyses, C-reactive protein, and procalcitonin. For empirical antibiotic therapy, the departments used (acyl-)aminopenicillins/β-lactamase inhibitors, cephalosporins, or combinations of (acyl-)aminopenicillins/β-lactamase inhibitors with fluoroquinolones or carbapenems. CONCLUSIONS This survey reveals a diagnostic ambiguity regarding SCI-AP despite the awareness of CDC-criteria and established SOPs. Heterogeneous clinical practice is encouraging the development of disease-specific guidelines for diagnosis and management of SCI-AP.
Collapse
Affiliation(s)
- Claudia Druschel
- Department of Neurology with Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
- Clinical and Experimental Spinal Cord Injury Research (Neuroparaplegiology), Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Orthopaedic and Trauma Surgery, Universitätsklinikum Carl-Gustav Carus, Dresden, Germany
| | - Ramin R Ossami Saidy
- Department of Neurology with Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
- Clinical and Experimental Spinal Cord Injury Research (Neuroparaplegiology), Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Ulrike Grittner
- Institute of Biometry and Clinical Epidemiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Claus P Nowak
- Institute of Biometry and Clinical Epidemiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Andreas Meisel
- Department of Neurology with Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- NeuroCure Clinical Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Klaus-Dieter Schaser
- Department of Orthopaedic and Trauma Surgery, Universitätsklinikum Carl-Gustav Carus, Dresden, Germany
| | - Andreas Niedeggen
- Treatment Centre for Spinal Cord Injuries, Trauma Hospital Berlin, Berlin, Germany
| | - Thomas Liebscher
- Department of Neurology with Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
- Clinical and Experimental Spinal Cord Injury Research (Neuroparaplegiology), Charité-Universitätsmedizin Berlin, Berlin, Germany
- Treatment Centre for Spinal Cord Injuries, Trauma Hospital Berlin, Berlin, Germany
| | - Marcel A Kopp
- Department of Neurology with Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany.
- Clinical and Experimental Spinal Cord Injury Research (Neuroparaplegiology), Charité-Universitätsmedizin Berlin, Berlin, Germany.
- Berlin Institute of Health (BIH), Berlin, Germany.
- Berlin Institute of Health, QUEST-Center for Transforming Biomedical Research, Berlin, Germany.
| | - Jan M Schwab
- Department of Neurology with Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
- Clinical and Experimental Spinal Cord Injury Research (Neuroparaplegiology), Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Neurology, Spinal Cord Medicine (Paraplegiology), The Neurological Institute, The Ohio State University, Wexner Medical Center, Columbus, OH, USA
- Belford Center for Spinal Cord Injury, Departments of Neuroscience and Physical Medicine and Rehabilitation, The Neurological Institute, The Ohio State University, Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
40
|
Carpenter RS, Marbourg JM, Brennan FH, Mifflin KA, Hall JCE, Jiang RR, Mo XM, Karunasiri M, Burke MH, Dorrance AM, Popovich PG. Spinal cord injury causes chronic bone marrow failure. Nat Commun 2020; 11:3702. [PMID: 32710081 PMCID: PMC7382469 DOI: 10.1038/s41467-020-17564-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 07/01/2020] [Indexed: 12/15/2022] Open
Abstract
Spinal cord injury (SCI) causes immune dysfunction, increasing the risk of infectious morbidity and mortality. Since bone marrow hematopoiesis is essential for proper immune function, we hypothesize that SCI disrupts bone marrow hematopoiesis. Indeed, SCI causes excessive proliferation of bone marrow hematopoietic stem and progenitor cells (HSPC), but these cells cannot leave the bone marrow, even after challenging the host with a potent inflammatory stimulus. Sequestration of HSPCs in bone marrow after SCI is linked to aberrant chemotactic signaling that can be reversed by post-injury injections of Plerixafor (AMD3100), a small molecule inhibitor of CXCR4. Even though Plerixafor liberates HSPCs and mature immune cells from bone marrow, competitive repopulation assays show that the intrinsic long-term functional capacity of HSPCs is still impaired in SCI mice. Together, our data suggest that SCI causes an acquired bone marrow failure syndrome that may contribute to chronic immune dysfunction.
Collapse
Affiliation(s)
- Randall S Carpenter
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH, USA
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH, USA
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA
| | - Jessica M Marbourg
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH, USA
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH, USA
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA
| | - Faith H Brennan
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH, USA
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA
| | - Katherine A Mifflin
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH, USA
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA
| | - Jodie C E Hall
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH, USA
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA
| | - Roselyn R Jiang
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH, USA
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA
| | - Xiaokui M Mo
- Center for Biostatistics and Bioinformatics, The Ohio State University, Columbus, OH, USA
| | - Malith Karunasiri
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Matthew H Burke
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Adrienne M Dorrance
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Division of Hematology, The Ohio State University, Columbus, OH, USA
| | - Phillip G Popovich
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA.
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH, USA.
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
41
|
Person JM, Welch BA, Spann RA, Harris KK, Pride Y, Tucci MA, Taylor EB, Grayson BE. Immuno-hematologic parameters following rodent spinal cord contusion are negatively influenced by high-fat diet consumption. J Neuroimmunol 2020; 343:577226. [PMID: 32247229 DOI: 10.1016/j.jneuroim.2020.577226] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/19/2020] [Accepted: 03/21/2020] [Indexed: 12/15/2022]
Abstract
Spinal cord injury (SCI) results in perturbations to the immune system leading to increased infection susceptibility. In parallel, the consumption of high-fat diets (HFD) leads to a chronic inflammation in circulation and body tissues. We investigated the impact of 16 weeks of HFD on chronically-injured rats. SCI rats under both chow and HFD showed peripheral leukocyte changes that include reduced percentages of total, helper and cytotoxic T, and natural killer cells. Expression of immune-related genes in the spleen and thymus reflected the impact of both chronic injury and diet. Changes to the immune system following SCI are adversely impacted by HFD consumption.
Collapse
Affiliation(s)
- Jon M Person
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS 39216, United States of America
| | - Bradley A Welch
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS 39216, United States of America
| | - Redin A Spann
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS 39216, United States of America
| | - Kwamie K Harris
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS 39216, United States of America
| | - Yilianys Pride
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS 39216, United States of America
| | - Michelle A Tucci
- Department of Anesthesiology, University of Mississippi Medical Center, Jackson, MS 39216, United States of America
| | - Erin B Taylor
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, United States of America
| | - Bernadette E Grayson
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS 39216, United States of America; Department of Anesthesiology, University of Mississippi Medical Center, Jackson, MS 39216, United States of America.
| |
Collapse
|
42
|
Vijapur SM, Yang Z, Barton DJ, Vaughan L, Awan N, Kumar RG, Oh BM, Berga SL, Wang KK, Wagner AK. Anti-Pituitary and Anti-Hypothalamus Autoantibody Associations with Inflammation and Persistent Hypogonadotropic Hypogonadism in Men with Traumatic Brain Injury. J Neurotrauma 2020; 37:1609-1626. [PMID: 32111134 DOI: 10.1089/neu.2019.6780] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Traumatic brain injury (TBI) and can lead to persistent hypogonadotropic hypogonadism (PHH) and poor outcomes. We hypothesized that autoimmune and inflammatory mechanisms contribute to PHH pathogenesis. Men with moderate-to-severe TBI (n = 143) were compared with healthy men (n = 39). The TBI group provided blood samples 1-12 months post-injury (n = 1225). TBI and healthy control (n = 39) samples were assayed for testosterone (T) and luteinizing hormone (LH) to adjudicate PHH status. TBI samples 1-6 months post-injury and control samples were assayed for immunoglobulin M (IgM)/immunoglobulin G (IgG) anti-pituitary autoantibodies (APA) and anti-hypothalamus autoantibodies (AHA). Tissue antigen specificity for APA and AHA was confirmed via immunohistochemistry (IHC). IgM and IgG autoantibodies for glial fibrillary acid protein (GFAP) (AGA) were evaluated to gauge APA and AHA production as a generalized autoimmune response to TBI and to evaluate the specificity of APA and AHA to PHH status. An inflammatory marker panel was used to assess relationships to autoantibody profiles and PHH status. Fifty-one men with TBI (36%) had PHH. An age-related decline in T levels by both TBI and PHH status were observed. Injured men had higher APA IgM, APA IgG, AHA IgM, AHA IgG, AGA IgM, and AGA IgG than controls (p < 0.0001 all comparisons). However, only APA IgM (p = 0.03) and AHA IgM (p = 0.03) levels were lower in the PHH than in the non-PHH group in multivariate analysis. There were no differences in IgG levels by PHH status. Multiple inflammatory markers were positively correlated with IgM autoantibody production. PHH was associated with higher soluble tumor-necrosis-factor receptors I/II, (sTNFRI, sTNFRII), regulated on activation, normal T-cell expressed and secreted (RANTES) and soluble interleukin-2-receptor-alpha (sIL-2Rα) levels. Higher IgM APA, and AHA, but not AGA, in the absence of PHH may suggest a beneficial or reparative role for neuroendocrine tissue-specific IgM autoantibody production against PHH development post-TBI.
Collapse
Affiliation(s)
- Sushupta M Vijapur
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Zhihui Yang
- Brain Rehabilitation Research Center, Malcom Randall VA Medical Center, North Florida / South Georgia Veterans Health System, Gainesville, Florida, USA.,Department of Emergency Medicine, Psychiatry and Neuroscience, University of Florida, Gainesville, Florida, USA
| | - David J Barton
- Department of Emergency Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Leah Vaughan
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Nabil Awan
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Raj G Kumar
- Mount Sinai, Icahn School of Medicine, New York, New York, USA
| | - Byung-Mo Oh
- Department of Rehabilitation Medicine, Seoul National University, Seoul, South Korea
| | - Sarah L Berga
- Department of Obstetrics and Gynecology, University of Utah Health Sciences Center, Salt Lake City, Utah, USA
| | - Kevin K Wang
- Department of Emergency Medicine, Psychiatry and Neuroscience, University of Florida, Gainesville, Florida, USA.,Department of Emergency Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Amy K Wagner
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Clinical and Translational Science Institute, University of Pittsburgh, Pennsylvania, USA
| |
Collapse
|
43
|
Jacob I, Rangappa P, Thimmegowda LC, Rao K. A Study of Multidrug-Resistant, Colistin-Only-Sensitive Infections in Intubated and Mechanically Ventilated Patients Over 2 Years. J Glob Infect Dis 2020; 12:5-10. [PMID: 32165795 PMCID: PMC7045758 DOI: 10.4103/jgid.jgid_179_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 05/09/2019] [Accepted: 01/27/2020] [Indexed: 11/04/2022] Open
Abstract
Background and Aims Multidrug-resistant, Gram-negative infections are increasingly common in the intensive care unit (ICU). This study compares the occurrence and outcome of colistin-only-sensitive (COS) infections among mechanically ventilated patients at a tertiary hospital ICU. Methods The study included adult patients admitted over a period of 2 years, who were intubated and mechanically ventilated for more than 48 h. They were divided into two groups, those with COS infections and those without, and their GCS and Acute Physiology and Chronic Health Evaluation II (APACHE II) scores, ICU length of stay, leukocyte count, and mortality were compared. COS patients were divided into neurosurgery, neurology, respiratory, and sepsis with bacteremia groups. The COS organisms in each group, their sources, ICU length of stay, ventilator-free days, and mortality were analyzed. Results Three hundred and one patients were selected, of whom 41 (13.6%) had COS infections. COS patients had a longer ICU length of stay than non-COS patients (P = 0.001) but comparable APACHE II and GCS scores, leukocyte count, and mortality. The sepsis group accounted for 8 out of 15 (53%) deaths among COS patients (P = 0.03). Acinetobacter baumannii accounted for 61% of the COS infections, Klebsiella pneumonia: 24.4%, Pseudomonas aeruginosa: 12.2%, and Escherichia coli: 2.4%. Endotracheal secretion cultures accounted for 65.8% of COS isolates, urine cultures 17%, pus cultures 7.3%, and blood cultures 4.9%. ICU length of stay, ventilator-free days, and mortality were similar between each COS organism. Conclusion Intubated patients with multidrug-resistant, COS infections have a longer stay in ICU than non-COS patients. COS infections associated with bacteremia have high mortality.
Collapse
Affiliation(s)
- Ipe Jacob
- Department of Critical Care, Columbia Asia Referral Hospital, Bengaluru, Karnataka, India
| | - Pradeep Rangappa
- Department of Critical Care, Columbia Asia Referral Hospital, Bengaluru, Karnataka, India
| | - Lakshman C Thimmegowda
- Department of Critical Care, Columbia Asia Referral Hospital, Bengaluru, Karnataka, India
| | - Karthik Rao
- Department of Critical Care, Columbia Asia Referral Hospital, Bengaluru, Karnataka, India
| |
Collapse
|
44
|
Cancer immunotherapy: Pros, cons and beyond. Biomed Pharmacother 2020; 124:109821. [PMID: 31962285 DOI: 10.1016/j.biopha.2020.109821] [Citation(s) in RCA: 404] [Impact Index Per Article: 80.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/12/2019] [Accepted: 12/18/2019] [Indexed: 12/22/2022] Open
Abstract
Cancer immunotherapy is an innovative treatment for tumors today. In various experiments and clinical studies, it has been found that immunotherapy does have incomparable advantages over traditional anti-tumor therapy, which can prolong progression-free survival (PFS) and overall survival (OS). However, immunotherapy has obvious complexity and uncertainty. Immunotherapy may also cause severe adverse reactions due to an overactive immune system. More effective and fewer adverse reactions immunological checkpoints are still under further exploration. This review gives an overview of recent developments in immunotherapy and indicates a new direction of tumor treatment through analyzing the pros and cons of immunotherapy coupled with keeping a close watch on the development trend of the immunotherapy future.
Collapse
|
45
|
Systemic inflammation in traumatic spinal cord injury. Exp Neurol 2019; 325:113143. [PMID: 31843491 DOI: 10.1016/j.expneurol.2019.113143] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 12/06/2019] [Accepted: 12/10/2019] [Indexed: 01/08/2023]
|
46
|
Sribnick EA, Weber MD, Hall MW. Innate immune suppression after traumatic brain injury and hemorrhage in a juvenile rat model of polytrauma. J Neuroimmunol 2019; 337:577073. [PMID: 31670063 DOI: 10.1016/j.jneuroim.2019.577073] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 09/16/2019] [Accepted: 09/22/2019] [Indexed: 12/20/2022]
Abstract
Traumatic injury in children is known to cause immune suppression. Polytrauma involving a traumatic brain injury (TBI) may increase this degree of immune suppression, which increases the risk of developing nosocomial infections, potentially causing secondary brain injury and worsening patient outcomes. Despite the high prevalence of polytrauma with TBI in children, mechanisms of immune suppression following such injuries remain poorly understood. Here, we used a combined animal injury model of TBI and hemorrhage to assess immune function after polytrauma. Pre-pubescent rats were injured using a prefrontal controlled cortical impact method and a controlled hemorrhage by femoral arteriotomy. Immune function was measured by whole blood ex-vivo tumor necrosis factor alpha production capacity following incubation with lipopolysaccharide, measuring the percentage of monocytes by flow cytometry, and by examining concentrations of plasma cytokines. The degree of brain injury was sufficient to produce deficits in spatial memory testing (Barnes maze). Both hemorrhage and TBI with hemorrhage (combined injury) reduced several of the measured plasma cytokines, as compared with TBI alone. The combined injury correlated with reduced concentration of monocytes and reduced tumor necrosis factor alpha production capacity at post-injury day 1. These results demonstrate that this animal model can be used to study post-injury immune suppression.
Collapse
Affiliation(s)
- Eric A Sribnick
- Department of Surgery, Division of Neurosurgery, Nationwide Children's Hospital, Columbus, OH, USA; Department of Neurosurgery, The Ohio State University College of Medicine, Columbus, OH, USA; Center for Clinical and Translation Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.
| | - Michael D Weber
- Center for Clinical and Translation Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.
| | - Mark W Hall
- Center for Clinical and Translation Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, Division of Critical Care, Nationwide Children's Hospital, Columbus, OH, USA.
| |
Collapse
|
47
|
Preece J, Haynes A, Gupta S, Becknell B, Ching C. Implications of Bacteriuria in Myelomeningocele Patients at Time of Urodynamic Testing. Top Spinal Cord Inj Rehabil 2019; 25:241-247. [PMID: 31548791 DOI: 10.1310/sci2503-241] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Objective: To identify those myelomeningocele (MMC) patients at risk for post-urodynamic study (UDS) complications. We hypothesized that patients who manage their bladder with clean intermittent catheterization (CIC) would have a greater risk of post-instrumentation complications due to higher rates of bacteriuria compared to those who freely void (FV). Design/Methods: Urine was collected from patients with MMC without augmentation cystoplasty undergoing routine renal ultrasound or urodynamic study (UDS). Samples were divided into those with bacteriuria (urine culture ≥10,000 colony-forming units) and those without. Post-UDS complications were evaluated and compared between CIC and FV patients. Results: A total of 91 urine samples from 82 total MMC patients were included for evaluation. Significantly more patients on CIC than those who FV had bacteriuria (67% vs 33%, p = .0457). From these urine samples, 54 were obtained at time of UDS of which 45 were from patients on CIC and 9 from FV patients. More patients on CIC had bacteriuria at the time of UDS than those who FV (60% vs 33%, respectively), but this did not reach significance (p = .1416). No patient with bacteriuria on CIC had a complication after UDS while one FV patient with bacteriuria developed post-UDS pyelonephritis. Conclusion: MMC patients with bacteriuria on CIC did not have post-UDS complications. Patients with bacteriuria who FV may be at particular risk for post-instrumentation UTI, providing guidance as to which MMC patients should undergo urine testing prior to UDS in order to prevent post-instrumentation pyelonephritis.
Collapse
Affiliation(s)
- Janae Preece
- Department of Urology, Children's Hospital of Michigan, Detroit, Michigan
| | - Andria Haynes
- Division of Urology, Nationwide Children's Hospital, Columbus, Ohio
| | - Sudipti Gupta
- Division of Urology, Nationwide Children's Hospital, Columbus, Ohio.,Center for Clinical and Translational Research, Nationwide Children's Hospital, Columbus, Ohio
| | - Brian Becknell
- Center for Clinical and Translational Research, Nationwide Children's Hospital, Columbus, Ohio.,Division of Nephrology, Nationwide Children's Hospital, Columbus, Ohio
| | - Christina Ching
- Division of Urology, Nationwide Children's Hospital, Columbus, Ohio.,Center for Clinical and Translational Research, Nationwide Children's Hospital, Columbus, Ohio
| |
Collapse
|
48
|
Stampas A, Dominick E, Zhu L. Evaluation of functional outcomes in traumatic spinal cord injury with rehabilitation-acquired urinary tract infections: A retrospective study. J Spinal Cord Med 2019; 42:579-585. [PMID: 29611464 PMCID: PMC6758698 DOI: 10.1080/10790268.2018.1452389] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
Abstract
OBJECTIVE The objective of this study was to evaluate the impact of urinary tract infections (UTIs) acquired during acute inpatient traumatic Spinal Cord Injury (tSCI) rehabilitation on Functional Independence Measure (FIM) gains. DESIGN Retrospective chart review of consecutive patients with tSCI admitted to an acute rehabilitation facility from 2007-2012. The primary outcome was FIM scores and the association with UTI. RESULTS The sample included 110 patients and 70 acquired UTIs. No demographic differences were observed between groups with and without UTI. Those with UTIs had significantly lower FIM motor scores, on admission, discharge, and gain, as well as lower FIM efficiency and longer lengths of stay compared to those without a UTI. Recurrence of UTI was associated with increased length of stay, but did not impact FIM motor gains. CONCLUSIONS There was a statistically significant correlation between acquired UTIs and lower FIM motor scores on admission, gain, and discharge in tSCI rehabilitation. The correlation of UTI and decreased FIM gains are similar to other neurorehabilitation populations with UTI occurrence. UTIs are an important negative variable when measuring functional outcomes in rehabilitation. Further prospective studies should be performed to investigate this correlation.
Collapse
Affiliation(s)
- Argyrios Stampas
- Department of PM&R University of Texas Health Science Center at Houston, TIRR Memorial Hermann, Houston, Texas, USA,Correspondence to: Argy Stampas, MD, TIRR Memorial Hermann, 1333 Moursund St., Rm 165.6D, Houston, Texas 77030, USA.
| | | | - Liang Zhu
- Center for Clinical and Translational Sciences, University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
49
|
Hong J, Chang A, Liu Y, Wang J, Fehlings MG. Incomplete Spinal Cord Injury Reverses the Level-Dependence of Spinal Cord Injury Immune Deficiency Syndrome. Int J Mol Sci 2019; 20:ijms20153762. [PMID: 31374824 PMCID: PMC6695842 DOI: 10.3390/ijms20153762] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 07/29/2019] [Accepted: 07/31/2019] [Indexed: 11/30/2022] Open
Abstract
Spinal cord injury (SCI) is associated with an increased susceptibility to infections, such as pneumonia, which is the leading cause of death in these patients. This phenomenon is referred to as SCI immune deficiency syndrome (SCI-IDS), and has been shown to be more prevalent after high-level transection in preclinical SCI models. Despite the high prevalence of contusion SCIs, the effects of this etiology have not been studied in the context of SCI-IDS. Compared to transection SCIs, which involve a complete loss of supraspinal input and lead to the disinhibition of spinally-generated activity, contusion SCIs may cause significant local deafferentation, but only a partial disruption of sympathetic tone below the level of injury. In this work, we investigate the effects of thoracic (T6-7) and cervical (C6-7) moderate–severe contusion SCIs on the spleen by characterizing splenic norepinephrine (NE) and cortisol (CORT), caspase-3, and multiple inflammation markers at 3- and 7-days post-SCI. In contrary to the literature, we observe an increase in splenic NE and CORT that correspond to an increase in caspase-3 after thoracic SCI relative to cervical SCI. Further, we found differences in expression of leptin, eotaxin, IP-10, and IL-18 that implicate alterations in splenocyte recruitment and function. These results suggest that incomplete SCI drastically alters the level-dependence of SCI-IDS.
Collapse
Affiliation(s)
- James Hong
- Institute for Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
- Krembil Research Institute, Toronto Western Hospital, Toronto, ON M5T 0S8, Canada
| | - Alex Chang
- Krembil Research Institute, Toronto Western Hospital, Toronto, ON M5T 0S8, Canada
| | - Yang Liu
- Krembil Research Institute, Toronto Western Hospital, Toronto, ON M5T 0S8, Canada
| | - Jian Wang
- Krembil Research Institute, Toronto Western Hospital, Toronto, ON M5T 0S8, Canada
| | - Michael G Fehlings
- Krembil Research Institute, Toronto Western Hospital, Toronto, ON M5T 0S8, Canada.
- Division of Neurosurgery, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
50
|
Chio JCT, Wang J, Badner A, Hong J, Surendran V, Fehlings MG. The effects of human immunoglobulin G on enhancing tissue protection and neurobehavioral recovery after traumatic cervical spinal cord injury are mediated through the neurovascular unit. J Neuroinflammation 2019; 16:141. [PMID: 31288834 PMCID: PMC6615094 DOI: 10.1186/s12974-019-1518-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 06/11/2019] [Indexed: 12/30/2022] Open
Abstract
Background Spinal cord injury (SCI) is a condition with few effective treatment options. The blood-spinal cord barrier consists of pericytes, astrocytes, and endothelial cells, which are collectively termed the neurovascular unit. These cells support spinal cord homeostasis by expressing tight junction proteins. Physical trauma to the spinal cord disrupts the barrier, which leads to neuroinflammation by facilitating immune cell migration to the damaged site in a process involving immune cell adhesion. Immunosuppressive strategies, including methylprednisolone (MPSS), have been investigated to treat SCI. However, despite some success, MPSS has the potential to increase a patient’s susceptibility to wound infection and impaired wound healing. Hence, immunomodulation may be a more attractive approach than immunosuppression. Approved for modulating neuroinflammation in certain disorders, including Guillain-Barre syndrome, intravenous administration of human immunoglobulin G (hIgG) has shown promise in the setting of experimental SCI, though the optimal dose and mechanism of action remain undetermined. Methods Female adult Wistar rats were subjected to moderate-severe clip compression injury (35 g) at the C7-T1 level and randomized to receive a single intravenous (IV) bolus of hIgG (0.02, 0.2, 0.4, 1, 2 g/kg), MPSS (0.03 g/kg), or control buffer at 15 min post-SCI. At 24 h and 6 weeks post-SCI, molecular, histological, and neurobehavioral effects of hIgG were analyzed. Results At 24 h post-injury, human immunoglobulin G co-localized with spinal cord pericytes, astrocytes, and vessels. hIgG (2 g/kg) protected the spinal cord neurovasculature after SCI by increasing tight junction protein expression and reducing inflammatory enzyme expression. Improvements in vascular integrity were associated with changes in spinal cord inflammation. Interestingly, hIgG (2 g/kg) increased serum expression of inflammatory cytokines and co-localized (without decreasing protein expression) with spinal cord vascular cell adhesion molecule-1, a protein used by immune cells to enter into inflamed tissue. Acute molecular benefits of hIgG (2 g/kg) led to greater tissue preservation, functional blood flow, and neurobehavioral recovery at 6 weeks post-SCI. Importantly, the effects of hIgG (2 g/kg) were superior to control buffer and hIgG (0.4 g/kg), and comparable with MPSS (0.03 g/kg). Conclusions hIgG (2 g/kg) is a promising therapeutic approach to mitigate secondary pathology in SCI through antagonizing immune cell infiltration at the level of the neurovascular unit.
Collapse
Affiliation(s)
- Jonathon Chon Teng Chio
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard Avenue, 7KD-430, Toronto, Ontario, M5T 2S8, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.,Spinal Program, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Jian Wang
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard Avenue, 7KD-430, Toronto, Ontario, M5T 2S8, Canada
| | - Anna Badner
- Sue and Bill Gross Stem Cell Research Centre, University of California, 845 Health Sciences Road, Irvine, CA, 92617, USA
| | - James Hong
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard Avenue, 7KD-430, Toronto, Ontario, M5T 2S8, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.,Spinal Program, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | | | - Michael G Fehlings
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard Avenue, 7KD-430, Toronto, Ontario, M5T 2S8, Canada. .,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada. .,Spinal Program, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada. .,University of Toronto, Toronto, Ontario, Canada. .,Gerry and Tootsie Halbert Chair in Neural Repair and Regeneration, University of Toronto, Toronto, Canada. .,Krembil Neuroscience Program, Toronto Western Hospital, University Health Network, 399 Bathurst Street, Toronto, Ontario, M5T 2S8, Canada.
| |
Collapse
|