Reducing Inflammation by Targeting Lymphocyte Expression in Relapse Remitting Multiple Sclerosis
Author: Virginia Buracioc
Neuroscience In Review
Introduction
Multiple sclerosis (MS) is an inflammatory, autoimmune-mediated neurological disease that affects the central nervous system (CNS). MS leads to severe cognitive, physical, and neurological issues in approximately 2.5 million young adults worldwide (Jelcic, I. et al., 2018). In addition, 15-30% of adults suffering from MS develop a relapsing-remitting (RR) course, manifested in neurological, physiological, sensory, and motor impairments which progress with intervals of acute recovery and remission (Kunkl et al., 2020). Current research suggests that
50 % of patients with RRMS progress into chronic secondary disability over a vacillating period. Though the exact mechanism of MS development is unknown, research focuses on determining potential MS causes and triggers of its progression (Kunkl et al., 2020).
Generally, MS targets the CNS by destroying the myelinated axons. Due to axon demyelination, patients develop a variety of visual, motor, sensory, autonomic, and neurocognitive impairments (Jelcic, I. et al., 2018). Although the pathogenesis of MS is still unclear, many studies support a conjugated effect in its progression. Both environmental and genetic predisposition contribute to developing its degenerative course (Ghasemi N. et al., 2017).) Potential exposure to environmental pathogens, infectious agents, viruses, smoking, and other factors stimulates the initiation of a negative cascade of events in the immune system resulting in myelin sheath demyelination and, ultimately, neuronal failure (Ghasemi, N. et al., 2017). In addition, studies suggest the intervention of T helper (CD4+ T) cells in conjugation with viral antigens initiate and progress inflammatory cytokine signaling (Fransen NL et al., 2020). Initiation of specific cytokines such as (IL)-12, IL-23, and IL-4 induce T cell phenotype differentiation which triggers additional innate and adaptive immunity proinflammatory cytokines (IFNγ) to promote neuronal inflammation by suppressing Th2 differentiation (Ghasemi N et al.,2017). This literature review will focus on understanding the mechanism of MS progression through the T helper cell differentiation pathway and investigating an effective therapeutic for individuals suffering from RRMS.
The role of T helper cells in initiating cytokine signaling
Extensive research shows T helper (Th) cells play a crucial role in the pathophysiology of MS. Specifically, CD4+T recruitment across the blood-brain barrier (BBB) into the CNS releases specific cytokines that trigger progressive inflammation throughout the brain (Ghasemi, N. et al., 2017). T helper cells are white blood cells that are crucial in sustaining adaptive immunity. Helper T cells function when activated by antigen-presenting cells, which promote differentiation into effector helper subclasses such as Th1 or Th2 cells, further allowing recognition by cytokines (Alberts B et al., 2002). Cell differentiation into Th1 cells initiates interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α) secretion, which triggers macrophages to attack oligodendrocytes. Thus, promoting myelin destruction, apoptosis of infected cells, cellular debris, lesions, and inflammation in the CNS (Alberts B et al., 2002). However, differentiation into Th2 cells initiates activation of anti-inflammatory interleukins (IL-4 and IL-13) that combat inflammation caused by cell death thus, resulting in restoration and initiation of new oligodendrocytes to induce remyelination of damaged axons (Alberts B et al., 2002).
T helper cell differentiation promotes inflammation in MS patients
Studies suggest that increased levels of Th cells and cytokines found in CNS lesions and cerebrospinal fluid (CSF) of MS patients confirm lymphocyte extravasation across the BBB leading to exacerbated progression of MS (Kunkl et al., 2020) (Alberts B et al., 2002). These signaling factors influence astrocyte and microglial activation leading to BBB destruction, resulting in neuroinflammation and myelin damage (Alberts B et al., 2002). Several current hypotheses mention specific T lymphocyte interactions with antigen-presenting cells (APCs) at the beginning and further progression of MS (Ghasemi N et al., 2017). The predicted mechanism of action includes binding of pathogen molecules such as viral infections, bacterial lipopolysaccharides, and metabolic stress with toll receptors present on APCs, triggering interleukin (IL)-12, IL-23, and IL-4) cytokines (Ghasemi N et al.,2017). Such cytokines further induce differentiation of CD4+ T cells into different Th phenotypes (Th1, Th2/Th17), able to initiate the release of additional pro-inflammatory cytokines. This cytokine cascade includes Th1-produced pro-inflammatory cytokines responsive to innate and adaptive immunity (Ghasemi N et al., 2017). Pro-inflammatory cytokines such as Interferon-gamma (IFNγ) or type II interferon (TNF-α) suppress Th2 cell differentiation, encouraging inflammation. Furthermore, research suggests Th2 cells’ role in pathological inflammation is controlled by releasing IL-4 cytokines, which reduces inflammation by increasing M2 macrophage (Ghasemi N et al.,2017). In conclusion, CD4+T differentiation into subsets such as Th1, Th2, and Th17 induces cytokine release that further triggers inflammation. Recruitment of CD4+ T lymphocytes across the BBB into the CNS develops inflammation and oligodendrocyte damage throughout the brain by activating several proinflammatory cytokines (Ghasemi N et al., 2017).
The cytokine IFN-γ produced by the T cells and natural killer cells plays a critical role in autoimmune demyelination. Although the molecular mechanisms of how cytokines mediate destruction of oligodendrocytes, demyelination is believed to be facilitated by cell-mediated cytotoxicity and apoptosis (Balabanov et al., 2006, Traugott et al., 2001). Studies have shown that upon activation of IFN-γ, oligodendrocytes upregulate the expression of surface ligands and and major histocompatibility (MHC) class I molecules that promote cytotoxicity and ultimately cell death (Balabanov et al., 2006). Research on autoimmune-mediated demyelination has also illustrated the effect of IFN-γ on failed remyelination in RRMS. Studies have attributed inability of oligodendrocytes to remyelinate in demyelinated lesions is due in part to endoplasmic reticulum (ER) stress (Wensheng et al., 2006, Lin et al., 2005). Mediating ER stress could be a potential target in facilitating remyelination in demyelinated lesions of MS patients to reduce relapse of symptoms. Meanwhile, one effective MS treatment that decreases inflammation and thus reduces symptoms in RRMS patients is Natalizumab.
Natalizumab (NAT) short term treatment reduces inflammation in RRMS patients
Natalizumab (NAT) is an effective treatment in active relapsing-remitting multiple sclerosis (RRMS) patients. NAT inhibits the migration and extravasation of leukocytes across the CNS by binding to α4-integrin (Lakritz et al., 2016) (Mellergård et al., 2013). This treatment is encouraged for RRMS patients that maintain non-responsive nor effective reactions to current effective therapy such as first-line disease-modifying therapies (DMT) (Kappos et al., 2011). Therefore, allowing individuals with no-responsive behavior is an alternative option in combating RRMS and improving their quality of life. A study observed the effects of short-term NAT treatment on lymphocyte circulation in 40 RRMS patients taking a drug dosage of 30 mg weekly (Mellergård et al., 2013). The results showed an increased expression of NK cells and B cells, including improved T cell (CD4+/CD8+) sensitivity to antigens (Mellergård et al., 2013). NAT treatment showed decreased migration of harmful lymphocytes across the BBB into the CNS and reduced intrathecal inflammation (Lakritz et al., 2016) (Mellergård et al., 2013). Similar results are observed in a study performed over two years that tested the effects of Natalizumab treatment in 596 patients (Polman et al., 2006). The study compared the annual relapse rate (ARR) after NAT treatment and placebo administration of one year. The results found a significant reduction following NAT treatment compared to the placebo.
Moreover, the study observed a 59% reduction in relapse following a two-year NAT treatment (Polman et al., 2006). Ultimately, the study aligns with other studies suggesting a decrease of RRMS and a decrease in disease progression and lesion formation in MS patients. These findings indicate that NAT could be used as an effective long-term treatment in RRMS patients.
Natalizumab (NAT) long term treatment reduces relapse remission in MS patients
Long-term treatment focuses on determining the efficacy, safety, and life quality of RRMS patients prescribed NAT. Furthermore, MS research shows the effectiveness of NAT treatment in reducing RR rates in MS patients in long-term use. A study performed by an observational program (TOP) analyzed ARRs of MS patients following the NAT treatment over ten years (Butzkueven et al., 2019). Results suggest a significant reduction of ARR across all subjects despite initial baseline, age, or category classification. Individuals with an initial lower baseline (less recorded relapse in the year before treatment) experience significant lower ARRs when on NAT treatment (Butzkueven et al., 2019). Additionally, during the first year of treatment, ARR significantly decreased from an initial baseline score of 1.99 to 0.24 and remained at this rate for the duration of the 10-year treatment (Butzkueven et al., 2019).
A similar study observed treatment annualized relapse rates in RRMS patients also revealed significantly reduced ARR over five years of treatment (Butzkueven et al., 2014). Besides efficient decreased annual relapse rates, researchers noted progressive multifocal leukoencephalopathy (PML) as a common side effect in multiple MS patients, increasing with yearly NAT treatment exposure (Kleinschmidt-DeMasters et al., 2005, Butzkueven et al., 2019). PML is a progressive disorder that maintains a rapid degenerative course, leading to the destruction of the white matter, manifested as extensive lesions throughout the brain (Kleinschmidt-DeMasters et al., 2005). Although the mechanism of PML development in MS patients is unknown, research has indicated that PML develops in individuals with autoimmune disorders such as MS and with other immunosuppressive diseases such as HIV and lymphomas (Major et al., 2018). PML is caused by the reactivation of the John Cunningham (JC) virus that is unharmful in its dormant stages (Major et al., 2018). However, in MS patients taking long-term immunosuppressive medication such as Natalizumab leads to the reactivation of the virus. Consequently, the virus leads to severe inflammation and damage to oligodendrocytes in various parts of the brain. The development of PML in MS patients suggests further research is needed on the effects of NAT treatment in MS patients that receive long-term NAT treatment. In addition, studies should investigate the impact of α4-integrin inhibition in RRMS patients and how it might play a role in developing PML.
Conclusion
Multiple sclerosis is a severe, progressive disease that affects millions worldwide. MS progression results in inflammation, axon demyelination, and relapse-remission stages resulting in various progressive autonomic and neurocognitive impairments. Although the exact mechanism of disease initiation and development remains unclear, scientists suggest factors such as T helper cell differentiation, pathogens, and cytokine signaling participate in MS advancement. Although current MS treatment involves first-line disease-modifying therapies (DMT) to reduce relapse frequency and severity, DMTs are not effective in all RRMS patients. However, Natalizumab is a potential short and long-term treatment, showing effective potency in reducing yearly relapse rates and increasing life quality. Despite its effectiveness and efficiency in reducing relapse remission rates, NAT progresses with detrimental side effects such as PML disease, leading to progressive deterioration, extensive lesions, and destruction of white matter in RRMS patients. Therefore, further research is needed to investigate the progression of PML as a significant side effect of NAT long-term treatment. Furthermore, current research indicates additional long-term NAT treatment conduction to acquire further data on annual disease relapse rates, safety, efficiency, and life quality during and after treatment. Since MS involves several cell types that trigger an array of specific immune responses, future research should also investigate the interaction of glial and immune cell types and their role in the progression of MS. Specifically, the interaction between oligodendrocytes, pro-inflammatory and anti-inflammatory cytokines released by natural killer cells and T cells such as IFN-γ and other cytokines involved in demyelination should be further investigated. Since, studies have shown the effect of IFN-γ on reduced remyelination in response to ER stress, further research should explore how oligodendrocytes can mediate ER stress to remyelinate lesions in the brain.
[+] References
Alberts B, Johnson A, Lewis J, et al. Molecular Biology of the Cell. 4th edition. New York: Garland Science; 2002. Helper T Cells and Lymphocyte Activation. Available from: https://www.ncbi.nlm.nih.gov/books/NBK26827/
Butzkueven H., Kappos, L., Wiendl, H., Trojano, M., Spelman, T., Chang, I., Kasliwal, R., Jaitly, S., Campbell, N., Ho, P. R., Licata, S., & Tysabri Observational Program (TOP) Investigators (2020). Long-term safety and effectiveness of natalizumab treatment in clinical practice: 10 years of real-world data from the Tysabri Observational Program (TOP). Journal of neurology, neurosurgery, and psychiatry, 91(6), 660–668. https://doi.org/10.1136/jnnp-2019-322326
Butzkueven H, Kappos L, Pellegrini F, et al Efficacy and safety of natalizumab in multiple sclerosis: interim observational programme results Journal of Neurology, Neurosurgery & Psychiatry 2014;85:1190-1197
B.K. Kleinschmidt-DeMasters, M.D., and Kenneth L. Tyler, M.D. Progressive Multifocal Leukoencephalopathy Complicating Treatment with Natalizumab and Interferon Beta-1a for Multiple Sclerosis, July 28, 2005 N Engl J Med 2005; 353:369-374 doi: 10.1056/NEJMoa051782
Chris H. Polman, M.D., Paul W. O'Connor, M.D., Eva Havrdova, M.D., Michael Hutchinson, M.D., Ludwig Kappos, M.D., David H. Miller, M.D., J. Theodore Phillips, M.D., Ph.D., Fred D. Lublin, M.D., Gavin Giovannoni, M.D., Andrzej Wajgt, M.D., Martin Toal, M.B., M.F.P.M., Frances Lynn. A Randomized, Placebo-Controlled Trial of Natalizumab for Relapsing Multiple Sclerosis., March 2, 2006N Engl J Med 2006; 354:899-910 doi: 10.1056/NEJMoa044397
Ghasemi, N., Razavi, S., & Nikzad, E. (2017). Multiple Sclerosis: Pathogenesis, Symptoms, Diagnoses and Cell-Based Therapy. Cell journal, 19(1), 1–10. https://doi.org/10.22074/cellj.2016.4867
Jelcic, I., Al Nimer, F., Wang, J., Lentsch, V., Planas, R., Jelcic, I., Madjovski, A., Ruhrmann, S., Faigle, W., Frauenknecht, K., Pinilla, C., Santos, R., Hammer, C., Ortiz, Y., Opitz, L., Grönlund, H., Rogler, G., Boyman, O., Reynolds, R., Lutterotti, A., … Martin, R. (2018). Memory B Cells Activate Brain-Homing, Autoreactive CD4+ T Cells in Multiple Sclerosis. Cell, 175(1), 85–100.e23. https://doi.org/10.1016/j.cell.2018.08.011
Kappos L, Bates D, Edan G, Eraksoy M, Garcia-Merino A, Grigoriadis N, Hartung HP, Havrdová E, Hillert J, Hohlfeld R, Kremenchutzky M, Lyon-Caen O, Miller A, Pozzilli C, Ravnborg M, Saida T, Sindic C, Vass K, Clifford DB, Hauser S, Major EO, O'Connor PW, Weiner HL, Clanet M, Gold R, Hirsch HH, Radü EW, Sørensen PS, King J. Natalizumab treatment for multiple sclerosis: updated recommendations for patient selection and monitoring. Lancet Neurol. 2011 Aug;10(8):745-58. doi: 10.1016/S1474-4422(11)70149-1. PMID: 21777829.
Kunkl M, Frascolla S, Amormino C, Volpe E, Tuosto L. T Helper Cells: The Modulators of Inflammation in Multiple Sclerosis. Cells. 2020; 9(2):482. https://doi.org/10.3390/cells9020482
Lakritz, J. R., Thibault, D. M., Robinson, J. A., Campbell, J. H., Miller, A. D., Williams, K. C., & Burdo, T. H. (2016). α4-Integrin Antibody Treatment Blocks Monocyte/Macrophage Traffic to, Vascular Cell Adhesion Molecule-1 Expression in, and Pathology of the Dorsal Root Ganglia in an SIV Macaque Model of HIV-Peripheral Neuropathy. The American journal of pathology, 186(7), 1754–1761. https://doi.org/10.1016/j.ajpath.2016.03.007
Mellergård J, Edström M, Jenmalm MC, Dahle C, Vrethem M, et al. (2013) Increased B Cell and Cytotoxic NK Cell Proportions and Increased T Cell Responsiveness in Blood of Natalizumab-Treated Multiple Sclerosis Patients. PLOS ONE 8(12): e81685. https://doi.org/10.1371/journal.pone.0081685
Mellergård, J., Edström, M., Jenmalm, M. C., Dahle, C., Vrethem, M., & Ernerudh, J. (2013). Increased B cell and cytotoxic NK cell proportions and increased T cell responsiveness in blood of natalizumab-treated multiple sclerosis patients. PloS one, 8(12), e81685. https://doi.org/10.1371/journal.pone.0081685
Balabanov, R., Strand, K., Kemper, A., Lee, J. Y., & Popko, B. (2006). Suppressor of cytokine signaling 1 expression protects oligodendrocytes from the deleterious effects of interferon-gamma. The Journal of neuroscience : the official journal of the Society for Neuroscience, 26(19), 5143–5152. https://doi.org/10.1523/JNEUROSCI.0737-06.2006
Wensheng Lin, April Kemper, Jeffrey L. Dupree, Heather P. Harding, David Ron, Brian Popko, Interferon-γ inhibits central nervous system remyelination through a process modulated by endoplasmic reticulum stress, Brain, Volume 129, Issue 5, May 2006, Pages 1306–1318, https://doi.org/10.1093/brain/awl044
Lin W, Harding HP, Ron D, Popko B. ER Stress modulates the response of myelinating oligodendrocytes to the immune cytokine interferon-gamma. J Cell Biol 2005; 169: 603–12.
Major, E. O., Yousry, T. A., & Clifford, D. B. (2018). Pathogenesis of progressive multifocal leukoencephalopathy and risks associated with treatments for multiple sclerosis: a decade of lessons learned. The Lancet Neurology, 17(5), 467-480.
Traugott U (2001). Evidence for immunopathogenesis. In: Handbook of multiple sclerosis (Cook S, ed.) Ed 3 pp. 163–192. New York: Dekker.
[+] Other Work By Virginia Buracioc
Natural Killer Cells in Amyotrophic Lateral Sclerosis
Neuroanatomy
In this study the subgroup of familial ALS (fALS) disorder was investigated by studying the presence of the mutant superoxide dismutase 1 (SOD1) gene with respect to Natural killer (NK) cells in the death of motor neurons (11). This study included both ALS patients and SOD1 mice and concluded that the presence of NK cells was correlated with ALS symptoms and ALS markers such as damaged microglia and the suppression of protective cells.
Hyperexcitability of PV interneurons’ effects on memory and learning in AD
Neurophysiology
In a recent study, hippocampal inhibitory parvalbumin (PV) interneurons were investigated in relation to neuron network dysfunction and memory impairments in Alzheimer's disease (AD). This research suggests early treatment innervation is sufficient to prevent early amyloid-beta-induced hyperexcitability of interneurons, leading to spared memory deficiency in mice models expressing build-up amyloid deposits (APP/PS1 mice). Additionally, this study mentions early intervening treatment results in long-term memory improvement, increased hippocampal network activity, reduced amyloid plaque deposition, and delayed AD progression. This study investigates a novel approach in AD research by looking at the effects of inhibitory network dysfunction in AD pathogenesis allowing for a more diverse research study of AD increasing the possibility of effective therapeutics (Hijazi et al., 2020).