Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Dec 1.
Published in final edited form as: Curr Opin Pharmacol. 2022 Oct 4;67:102303. doi: 10.1016/j.coph.2022.102303

Systemic inflammation after spinal cord injury: A review of biological evidence, related health risks, and potential therapies

Gregory E Bigford 1, Eric Garshick 2,3
PMCID: PMC9929918  NIHMSID: NIHMS1867920  PMID: 36206621

Abstract

Individuals with chronic traumatic spinal cord injury (SCI) develop progressive multi-system health problems that result in clinical illness and disability. Systemic inflammation is associated with many of the common medical complications and acquired diseases that accompany chronic SCI, suggesting that it contributes to a number of comorbid pathological conditions. However, many of the mechanisms that promote persistent systemic inflammation and its consequences remain ill-defined. This review describes the significant biological factors that contribute to systemic inflammation, major organ systems affected, health risks, and the potential treatment strategies. We aim to highlight the need for a better understanding of inflammatory processes, and to establish appropriate strategies to address inflammation in SCI.

Introduction

Traumatic spinal cord injury (SCI) incites an immediate, adaptive inflammatory response characterized by activation of host defense against the tissue damage. Acute inflammation occurs in the spinal cord injury micro-environment at the time of injury, but residual systemic inflammation can continue indefinitely [1] in individuals living with chronic SCI. Chronic SCI is generally defined as ≥ 1-year post-injury, wherein there is stabilization of residual neurological function and a general physiological homeostasis of body composition, hormonal profile, and inflammatory environment [2]. Sensorimotor deficits are an obvious consequences of SCI, however, the spinal cord also plays a critical role in coordinating bodily functions [3] via complex bidirectional communication between the autonomic, endocrine, and immune systems [4,5], which may also be damaged after SCI and result in widespread dysfunction in multiple organ systems [6-8]. For example, with long-term injury, physical limitations related to movement, musculoskeletal activity and weight-bearing contribute to deleterious changes in body composition, including rapid and long-term declines in metabolically active muscle-mass [9-17] and bone [18-24], and significant increases in central adiposity [25-27]. Recurrent and chronic infection in the skin attributable to pressure ulcers [28] and urinary tract infections due to bladder dysfunction and catheter use are common following spinal cord injury [29,30]. These pathophysiological changes are all associated with an inflammatory response [5,6] which appears to promote systemic inflammation in individuals with chronic SCI.

Several recent reviews have previously described chronic systemic inflammation [5,31,32], immune [5] and organ dysfunction [6] associated with SCI. Our focus here is on evidence from human population and epidemiological studies on systemic inflammation in chronic SCI, although we highlight emerging evidence from novel preclinical research. We review the biological factors that contribute to chronic inflammation systemically and in peripheral organs, clinical manifestations, and potential countermeasures to inflammatory-induced pathologies after SCI.

General biomarkers of systemic inflammation

Chronic inflammatory conditions are associated with the prolonged release of inflammatory mediators and the activation of harmful signal–transduction pathways that contribute to aging-related phenotypes and disease development [33] such as cardiovascular disease [34], type II diabetes [35], and many cancers [36]. A multitude of circulating “inflammatory and immune” biomarkers, including cytokines—such as TNF-α, IL-1β, IL-6; acute-phase reactants—such as CRP; chemokines—such as IL-8 and MCP-1; and soluble immune factors, are commonly reported in epidemiologic and intervention studies to be associated with disease risk [37,38]. In severe cases, acute injury, trauma and infection, incite excessive production of cytokines in a so called pro-inflammatory “cytokine storm” [39,40], which are linked to chronic conditions described as “systemic inflammatory response syndrome” and “multi-organ dysfunction syndrome.” In these circumstances, the persistent elevation of cytokines is associated with long-term damaging effects on organ systems and the development of chronic disease [41]. Importantly, it is now well reported that severe acute SCI and an accompanying pro-inflammatory cytokine storm (Figure 1) incites such systemic and multi-organ inflammatory responses [42-46] where the neurological extent and severity of injury are closely associated with more advanced long-term health complications [6,47]. Both preclinical [48] and post-mortem human data [49] indicate that inflammation at the injury site persists into chronic phases of SCI, suggesting it may contribute to systemic inflammation.

Figure 1.

Figure 1

SCI-Induced acute inflammatory response at the injury site. Reactive Glia including astrocytes and microglia, and infiltrating immune cells including lymphocytes, macrophages, neutrophils that penetrate the damaged blood–spinal cord barrier, both release a myriad of pro-inflammatory cytokines and factors into the injury micro-environment resulting in detrimental neurological deficits.

Systemic inflammatory biomarkers and related health complications in chronic SCI

Both case reports and larger cohort studies provide evidence of a heightened pro-inflammatory state with chronic SCI [31,50,51]. In particular, CRP, an established clinical marker of systemic inflammation and cardiovascular disease risk [52,53] is elevated in individuals with chronic SCI [54-59]. A recent pooled-analysis of SCI-specific studies (n = 259), demonstrated that mean CRP levels accompanying SCI exceed high-risk criterion guidelines [51,60] for acute infection or chronic inflammatory diseases (such as cardiovascular disease). Importantly, CRP is considered a predictor of myocardial infarction, peripheral vascular disease, stroke, and sudden cardiac death [61], and as described by Nash et al. [51], CRP integration into global cardiovascular disease risk determination, worsens the hazard ratio for cardiometabolic syndrome in SCI subjects [51]. Another study in subjects with chronic SCI (n = 93), found that higher mean CRP was associated with fasting insulin resistance and severe dyslipidemia [55], both metabolic risk factors for cardiovascular disease.

Associations with several additional pro-inflammatory cytokines have also been reported with chronic SCI. Several studies report elevated levels of TNF-α [62,63], IL-6 [58,64], and IL-2 [62] in subjects with chronic SCI compared to uninjured controls, and with SCI, elevated levels of IL-6 are also positively correlated with vascular cellular adhesion molecule-1 (VCAM-1) and intracellular adhesion molecule-1 (IAM-1), indicators of vascular inflammation [58]. Interestingly, a similarly powered study indicated no differences compared to controls with respect to TNF-α and IL-6 [54]. Some studies aim to assess the contribution of infection/skin ulcers, age, level, extent, and duration of injury and other clinical factors to an SCI-related chronic inflammatory state. One recent report has studied this in depth with respect to CRP levels in chronic SCI. Goldstein et al. [65] found that higher CRP was associated with greater BMI, urinary catheter use, recent respiratory illness, and non-white race. They also found that mean CRP increased with decreasing mobility, and overall, these characteristics were more important than the level and completeness of injury. Nonetheless, collectively, CRP, TNF-α, and IL-6 are cytokines most notably reported as pro-atherogenic, and associated with other cardiometabolic risks including insulin resistance, depressed plasma HDL-C, and obesity [66-69], all of which are commonly observed in chronic SCI [51,70-72]. This is important as mortality due to chronical cardiovascular disease has become more common in chronic SCI [73-75]. An additional SCI-specific risk factor in patients with injury at or above the level of sympathetic outflow (T6) [76], is autonomic dysfunction and the clinical manifestation of autonomic dysreflexia, known to exacerbate inflammatory stress [77], potentially accelerating pro-atherogenic processes and cardiovascular disease [75,77,78].

Further epidemiological studies indicate that the chronic inflammatory milieu post-SCI is associated with an array of secondary health risks in many organs and systems including the respiratory, gastrointestinal (GI) and urinary systems. For example, several cross-sectional studies found a significant inverse association between plasma levels of CRP and IL-6 and indices of pulmonary dysfunction that included reduced forced expiratory volume (FEV) and forced vital capacity (FVC) [79,80]. Although the biological role of these cytokines in pulmonary dysfunction have not been directly studied, these observations may again be important, given the aforementioned relationship between cytokines and chronic organ failure, and that pulmonary complications remain among the most common causes of mortality in chronic SCI [73,74,81-83]. In addition, the impact of SCI on the GI system results in several complications of which motility issues and bowel management are most frequently reported [84-86]. More recently, gut dysbiosis has been implicated in SCI-mediated GI pathophysiology, which may be a novel source of inflammation. Bacterial-derived lipopolysaccharides (LPS) induces a low-grade inflammatory state associated with obesity, insulin resistance, diabetes and CVD [87,88], and short-chain fatty acids (SCFA), are known to play an important role in regulating inflammation and insulin resistance [89]. As reviewed by Noller et al. [31] one study in humans with SCI found a decrease in SCFA-producing bacteria which was associated with greater bowel dysfunction compared to controls [90]. Interestingly, there is also an association between asymptomatic bacteriuria and elevated levels of inflammatory and immune biomarkers [91] (although not CRP, TNF-α, or IL-6), and a recent report described differences in the urinary microbiome between subjects with SCI and non-disabled control, and with time since injury in the SCI cohort [92]. Taken together, these studies suggest that microbiome alterations with SCI may contribute to inflammatory stress in the GI and urinary system, although additional research is needed to understand whether changes in the microbiota in these systems are pathological and/or contribute to associated cardiometabolic risks.

One of the most severe consequences of SCI is a profound alteration in body composition typified by rapid and long-term declines in sub-lesional bone and muscle mass, and an increase in adiposity, which has resulted in the prevalence of obesity with chronic SCI [71], and is predictive for cardiometabolic syndrome and CVD [93,94]. Obesity has been identified as the most prevalent cardiometabolic disease component risk accompanying SCI [95], and co-exists with dyslipidemia, glucose intolerance, and insulin resistance [56,96-98], all being comorbidities that are strongly associated with a “pro-inflammatory phenotype” that accelerates atherogenesis and CVD [99,100]. There is increasing scientific evidence implicating “pro-inflammatory” adipose-derived cytokines–adipocytokines—as a principal source of systemic inflammatory factors in both chronic SCI and non-disabled populations [56,58,101-103]. In both SCI and nondisabled individuals meeting the clinical definition of obesity, serum levels of IL-6 were significantly higher than in non-obese individuals [56,104-106], and in SCI and obesity, a positive correlation was reported between IL-6 and fasting insulin levels [58,107]. Another study found that after SCI, the pro-inflammatory adipocytokines Plasminogen activator inhibitor-1 (PAI-1)—which is elevated in obesity and type 2 diabetes mellitus [108-111], and implicated in atherosclerosis [112]—is negatively correlated with both HDL-C (“good cholesterol”) and the anti-inflammatory adipocytokine adiponectin, suggesting PAI-1 may contribute to obesity mediated metabolic complications, albeit conjecture at this point. Additionally, a recent report found an inverse relationship between plasma leptin and reduced pulmonary function in chronic SCI [113], suggesting a link between alterations in body composition and the metabolic syndrome with post SCI-pulmonary function loss.

Inflammatory and immune-mediated pathology in chronic SCI

Inflammation is a component of the immune system response to SCI. Functional crosstalk occurs between the nervous and immune systems via the autonomic nervous system (ANS) and the hypothalamic–pituitary–adrenal (HPA) axis [1,114] and lymphoid tissues [115] such as the spleen, thymus, lymph nodes, adrenal gland and bone marrow [5,115,116]. Following SCI, neuro-endocrine and immune system dysfunction result in a “state of immunosuppression” [5,117] including functional deficits in macrophages, natural killer cells, neutrophils, and lymphocytes [4,115,118], having severe consequences such as susceptibility to infection [4,117,119,120]. In fact, as reviewed by Bloom et al. [32], infections are the leading cause of re-hospitalization and of mortality for persons with SCI, who are more than 80 times more likely to die of sepsis than uninjured persons [32]. Importantly, immune cells—most notably macrophages, NK cells, T and B cells—produce and secrete pro-inflammatory cytokines—such as TNF-α and a host of interleukins including IL-1β and IL-6 [121]—in response to infection, injury or trauma [32]. As such, there is both an elevated inflammatory state, and a state of immunosuppression accompanying SCI [5], where deleterious effects of chronic inflammation further exacerbates susceptibility to infection and reduction in wound-healing capabilities [122] by inciting suppressive immune cell phenotypes [32]. Therefore, it is possible that impaired immunity after SCI may contribute to the occurrence of urinary tract infections [62,123], pulmonary infection [124-126], and skin ulcer occurrence and complications [5,127]. Recent neuro-epidemiological studies (n > 1400) demonstrated that post-SCI infections are independent risk factors for poorer recovery, suggesting it is an independent confound for neurobiological rehabilitation [117,128,129], highlighting the complexity and interaction of an ‘uncontrolled’ pro-inflammatory state and immune impairment that accompanies chronic SCI.

Countermeasures to inflammation in chronic SCI

Rehabilitation strategies, or ‘lifestyle’ modifications, such as anti-inflammatory diets and physical activity paradigms have been directly tested in populations with chronic SCI. Several concurrent studies by Allison et al. [130-134] have reported that anti-inflammatory diets are sufficient to reduce several key markers of inflammation including interferon gamma (IFN-γ), TNF-α, IL-1β, and IL-6. Additionally, regular physical activity/training (vs. acute/single session exercise) is understood to have anti-inflammatory effects, particularly under conditions of cardiovascular/cardiometabolic diseases [135], and various exercise paradigms are established strategies to modulate the immune and inflammatory systems in the SCI population [31,136-139]. For example, studies in chronic SCI have demonstrated an inverse relationship between physical activity levels and the cytokines CRP and IL-6 [140,141], and reduced levels of IL-6 with FES cycling [141,142]. Several other population-specific reports found that a long-term arm-crank exercise program reduced systemic levels of TNF-α, IL-6 and the adipocytokine leptin [143], and disparate hand cycling modalities were effective in lowering CRP, IL-6, and CVD risk factors, including waist circumference and insulin resistance [144]. A recent case-series evaluating therapeutic intervention combining diet and exercise paradigms reported successful results in reducing major cardiometabolic risks including insulin resistance [145], albeit very limited in scale and inflammatory data were not reported. More novel approaches to physical activity in SCI include exoskeletal-associated-walking (EAW) [146-149] and home-based exercise programs [150,151], which have included metabolic profiling, although larger studies and more comprehensive results are also needed to directly evaluate their effect on inflammation and cardiometabolic outcomes.

Given their role in chronic inflammation and disease states, several anti-cytokine therapies targeting TNF-α, IL-1β, and IL-6 have been developed and used to treat inflammation in a number of diseases, including rheumatoid arthritis, Crohn’s disease, psoriasis, arthritis and lupus [152,153]. As extensively reviewed by Bloom et al. [32], many anti-inflammatory strategies have focused on acute SCI pathophysiology and neurological outcomes rather than on long-term outcomes. However, a recent study in an in-vivo model of SCI-induced inflammation and cardiovascular disease may help guide future clinical studies. Bigford et al. [154] directly examined the relationship and time-course between inflammatory cytokines and atherosclerotic disease after SCI in a mouse model, and found that SCI significantly increased disease burden, and several cytokines—including TNF-α, IL-1β, and IL-6—were associated with overall disease load [154]. Importantly, they report that NF-κB inhibition via Salsalate (non-acetylated salicylates) —shown to significantly reduce several inflammatory markers including TNFα and IL-6—attenuated overall inflammatory load and significantly reduced disease. These data highlight that SCI directly incites an accelerated trajectory of systemic inflammation and disease and the potential for anti-inflammatory therapy to mitigate pro-inflammatory and proatherogenic consequences of SCI. Although other studies using animal models have examined immunomodulators of inflammation-associated pathways, small molecule agonists/antagonists of inflammatory signals, and intracellular components of inflammatory machinery (Reviewed in the study by Sun et al. [6]), translational studies applied to human SCI have not yet amassed meaningful data. Although statins are used for cardiovascular risk reduction based on risk score and are indicated6 in persons with diabetes regardless of score [155], current guidelines do not include mention of chronic SCI as a chronic inflammatory risk-enhancing condition where early statin use can be considered.

Emerging targets of inflammations in preclinical studies

Previous evidence has shown that individuals with SCI have greater visceral adipose tissue (VAT) cross-sectional area (i.e., indicative of central obesity), associated with maladaptive metabolic profile, and predictive of impaired glucose tolerance, insulin resistance, and dyslipidemia [156,157]. Again, pre-clinical studies may help direct our understanding of underlying biological mechanisms that contribute to inflammation. For example, VAT and pancreas represent bioactive sites of pro-inflammatory processes mediated by macrophage and T cell activation of the NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome. Activation of the NLRP3 inflammasome is now understood as the mechanism of activation and release of the pro-inflammatory cytokines IL-1β and IL-18 [158], principal in the induction of obesity, insulin resistance, and impaired glucose metabolism [159-161]. A large literature has brought attention to inflammasome activation as an acute innate response to trauma in the CNS [162-164] including acute injury to peripheral lung tissue [165]. One report by Bigford et al. [44] examined NRLP3 inflammasome in both VAT and pancreas in a model of chronic SCI, reporting activation of the inflammasome complex, and subsequent activation of cytokines IL-1β and IL-18, suggesting the inflammasome contributes to chronically acquired low-grade inflammation associated with metabolic disorders and component CVD risk. As reviewed by Wang et al. [166] many therapeutics have been developed to target the NLRP3 inflammasome, with potential in the therapeutic treatment of chronic neurological diseases such as Parkinson and Alzheimer disease, as well as atherosclerosis. Further research is necessary to determine if the inflammasome is a viable target in clinical practice.

Conclusion and future perspective

Chronic SCI results in systemic inflammation, immunosuppression, and multiple organ dysfunction which, left unresolved, leads to an exacerbated maladaptive response of body systems [7,167-169], and increased morbidity and mortality due to chronically acquired diseases such as all-cause CVD [74,170] (Figure 2). Understanding relationships between factors associated with systemic inflammation in chronic SCI and between systemic inflammation, immunosuppression, and organ system function will provide insight into pathways of disease and guide new approaches to prevent chronically acquired diseases. Understanding these relationships will also provide insight into the rationale behind novel approaches to prevent illness, morbidity, and mortality in chronic SCI.

Figure 2.

Figure 2

Systemic and peripheral organ inflammation with chronic SCI. SCI results in long-term and unresolved neurological deficits that have chronic pathological consequences on body systems. Concomitant tissue and organ dysfunction can directly contribute to systemic inflammation and/or lead to major clinical health risks. These health risks further contribute to systemic inflammation resulting in a “feed-forward” system of inflammation. Progressive and unresolved health risks lead to the prevalence of chronic diseases. →downstream effect; → contributes to systemic inflammation; -- > hypothesized; VAT = Visceral Adipose Tissue.

Funding:

Dr. Garshick was supported by VA Rehabilitation Research and Development Merit Review Grants B6618R and I01 RX000792 from the U.S. Department of Veterans Affairs, Rehabilitation Research and Development Service; and NIH NIAMS Grant R01 AR059270. The contents do not represent the views of the U.S. Department of Veterans Affairs or the United States Government.

Footnotes

Conflict of interest statement

Nothing declared.

References

  • 1.Schwab JM, et al. : The paradox of chronic neuroinflammation, systemic immune suppression, autoimmunity after traumatic chronic spinal cord injury. Exp Neurol 2014, 258:121–129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Farkas GJ, et al. : Nutritional status in chronic spinal cord injury: a systematic review and meta-analysis. Spinal Cord 2019, 57:3–17. [DOI] [PubMed] [Google Scholar]
  • 3.Stein DM, et al. : Risk factors for organ dysfunction and failure in patients with acute traumatic cervical spinal cord injury. Neurocritical Care 2010, 13:29–39. [DOI] [PubMed] [Google Scholar]
  • 4.Cruse JM, et al. : Immune system-neuroendocrine dysregulation in spinal cord injury. Immunol Res 1996, 15:306–314. [DOI] [PubMed] [Google Scholar]
  • 5.Allison DJ, Ditor DS: Immune dysfunction and chronic inflammation following spinal cord injury. Spinal Cord 2015, 53:14–18. [DOI] [PubMed] [Google Scholar]
  • 6.Sun X, et al. : Multiple organ dysfunction and systemic inflammation after spinal cord injury: a complex relationship. J Neuroinflammation 2016, 13:260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Kemp BJ: What the rehabilitation professional and the consumer need to know. Phys Med Rehabil Clin 2005, 16:1–18 [vii]. [DOI] [PubMed] [Google Scholar]
  • 8.SaKM G: The state of aging and public health for people with spinal cord injury: lost in transition? Top Spinal Cord Inj Rehabil 2010, 15:1–10. [Google Scholar]
  • 9.Burnham R, et al. : Skeletal muscle fibre type transformation following spinal cord injury. Spinal Cord 1997, 35:86–91. [DOI] [PubMed] [Google Scholar]
  • 10.Castro MJ, et al. : Influence of complete spinal cord injury on skeletal muscle cross-sectional area within the first 6 months of injury. Eur J Appl Physiol Occup Physiol 1999, 80:373–378. [DOI] [PubMed] [Google Scholar]
  • 11.Castro MJ, et al. : Influence of complete spinal cord injury on skeletal muscle within 6 mo of injury. J Appl Physiol (1985) 1999, 86:350–358. [DOI] [PubMed] [Google Scholar]
  • 12.Gorgey AS, Dudley GA: Skeletal muscle atrophy and increased intramuscular fat after incomplete spinal cord injury. Spinal Cord 2007, 45:304–309. [DOI] [PubMed] [Google Scholar]
  • 13.Grimby G, et al. : Muscle fiber composition in patients with traumatic cord lesion. Scand J Rehabil Med 1976, 8:37–42. [PubMed] [Google Scholar]
  • 14.Lotta S, et al. : Morphometric and neurophysiological analysis of skeletal muscle in paraplegic patients with traumatic cord lesion. Paraplegia 1991, 29:247–252. [DOI] [PubMed] [Google Scholar]
  • 15.Modlesky CM, et al. : Assessment of skeletal muscle mass in men with spinal cord injury using dual-energy X-ray absorptiometry and magnetic resonance imaging. J Appl Physiol (1985) 2004, 96:561–565. [DOI] [PubMed] [Google Scholar]
  • 16.Round JM, et al. : Fibre areas and histochemical fibre types in the quadriceps muscle of paraplegic subjects. J Neurol Sci 1993, 116:207–211. [DOI] [PubMed] [Google Scholar]
  • 17.Scelsi R, et al. : Muscle fiber type morphology and distribution in paraplegic patients with traumatic cord lesion. Histochemical and ultrastructural aspects of rectus femoris muscle. Acta Neuropathol 1982, 57:243–248. [DOI] [PubMed] [Google Scholar]
  • 18.Battaglino RA, et al. : Spinal cord injury-induced osteoporosis: pathogenesis and emerging therapies. Curr Osteoporos Rep 2012, 10:278–285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Frey-Rindova P, et al. : Bone mineral density in upper and lower extremities during 12 months after spinal cord injury measured by peripheral quantitative computed tomography. Spinal Cord 2000, 38:26–32. [DOI] [PubMed] [Google Scholar]
  • 20.Garland DE, et al. : Osteoporosis after spinal cord injury. J Orthop Res 1992, 10:371–378. [DOI] [PubMed] [Google Scholar]
  • 21.Giangregorio L, McCartney N: Bone loss and muscle atrophy in spinal cord injury: epidemiology, fracture prediction, and rehabilitation strategies. J Spinal Cord Med 2006, 29:489–500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Maimoun L, et al. : Bone loss in spinal cord-injured patients: from physiopathology to therapy. Spinal Cord 2006, 44:203–210. [DOI] [PubMed] [Google Scholar]
  • 23.Qin W, Bauman WA, Cardozo C: Bone and muscle loss after spinal cord injury: organ interactions. Ann N Y Acad Sci 2010, 1211:66–84. [DOI] [PubMed] [Google Scholar]
  • 24.Zehnder Y, et al. : Long-term changes in bone metabolism, bone mineral density, quantitative ultrasound parameters, and fracture incidence after spinal cord injury: a cross-sectional observational study in 100 paraplegic men. Osteoporos Int 2004, 15:180–189. [DOI] [PubMed] [Google Scholar]
  • 25.Gorgey AS, Gater DR: A preliminary report on the effects of the level of spinal cord injury on the association between central adiposity and metabolic profile. Pharm Manag PM R 2011, 3:440–446. [DOI] [PubMed] [Google Scholar]
  • 26.Groah SL, et al. : Nutrient intake and body habitus after spinal cord injury: an analysis by sex and level of injury. J Spinal Cord Med 2009, 32:25–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Spungen AM, et al. : Factors influencing body composition in persons with spinal cord injury: a cross-sectional study. J Appl Physiol 2003, 95:2398–2407. [DOI] [PubMed] [Google Scholar]
  • 28.Kruger EA, et al. : Comprehensive management of pressure ulcers in spinal cord injury: current concepts and future trends. J Spinal Cord Med 2013, 36:572–585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Ronco E, et al. : Diagnostic criteria of urinary tract infection in male patients with spinal cord injury. Neurorehabilitation Neural Repair 2011, 25:351–358. [DOI] [PubMed] [Google Scholar]
  • 30.D’Hondt F, Everaert K: Urinary tract infections in patients with spinal cord injuries. Curr Infect Dis Rep 2011, 13:544–551. [DOI] [PubMed] [Google Scholar]
  • 31.Noller CM, Groah SL, Nash MS: Inflammatory stress effects on health and function after spinal cord injury. Top Spinal Cord Inj Rehabil 2017, 23:207–217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Bloom O, Herman PE, Spungen AM: Systemic inflammation in traumatic spinal cord injury. Exp Neurol 2020, 325, 113143. [DOI] [PubMed] [Google Scholar]
  • 33.Liu CH, et al. : Biomarkers of chronic inflammation in disease development and prevention: challenges and opportunities. Nat Immunol 2017, 18:1175–1180. [DOI] [PubMed] [Google Scholar]
  • 34.Madjid M, Fatemi O: Components of the complete blood count as risk predictors for coronary heart disease: in-depth review and update. Tex Heart Inst J 2013, 40:17–29. [PMC free article] [PubMed] [Google Scholar]
  • 35.McArdle MA, et al. : Mechanisms of obesity-induced inflammation and insulin resistance: insights into the emerging role of nutritional strategies. Front Endocrinol 2013, 4:52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Aggarwal BB: Inflammation, a silent killer in cancer is not so silent! Curr Opin Pharmacol 2009, 9:347–350. [DOI] [PubMed] [Google Scholar]
  • 37.Navarro SL, et al. : Factors associated with multiple biomarkers of systemic inflammation. Cancer Epidemiol Biomarkers Prev 2016, 25:521–531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Calder PC, et al. : A consideration of biomarkers to be used for evaluation of inflammation in human nutritional studies. Br J Nutr 2013, 109:S1–S34. [DOI] [PubMed] [Google Scholar]
  • 39.Makhija R, Kingsnorth AN: Cytokine storm in acute pancreatitis. J Hepatobiliary Pancreat Surg 2002, 9:401–410. [DOI] [PubMed] [Google Scholar]
  • 40.Ulloa L, Tracey KJ: The “cytokine profile”: a code for sepsis. Trends Mol Med 2005, 11:56–63. [DOI] [PubMed] [Google Scholar]
  • 41.Jaffer U, Wade RG, Gourlay T: Cytokines in the systemic inflammatory response syndrome: a review. HSR Proc Intensive Care Cardiovasc Anesth 2010, 2:161–175. [PMC free article] [PubMed] [Google Scholar]
  • 42.Anthony DC, Couch Y: The systemic response to CNS injury. Exp Neurol 2014, 258:105–111. [DOI] [PubMed] [Google Scholar]
  • 43.Bao F, et al. : Increased oxidative activity in human blood neutrophils and monocytes after spinal cord injury. Exp Neurol 2009, 215:308–316. [DOI] [PubMed] [Google Scholar]
  • 44.Bigford GE, et al. : Neuroendocrine and cardiac metabolic dysfunction and NLRP3 inflammasome activation in adipose tissue and pancreas following chronic spinal cord injury in the mouse. ASN Neuro 2013, 5:243–255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Bigford GE, et al. : Alterations in mouse hypothalamic adipokine gene expression and leptin signaling following chronic spinal cord injury and with advanced age. PLoS One 2012, 7, e41073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Popovich PG, et al. : Alterations in immune cell phenotype and function after experimental spinal cord injury. J Neurotrauma 2001, 18:957–966. [DOI] [PubMed] [Google Scholar]
  • 47.Kesani AK, et al. : Systemic inflammatory response syndrome in patients with spinal cord injury: does its presence at admission affect patient outcomes? Clinical article. J Neurosurg Spine 2014, 21:296–302. [DOI] [PubMed] [Google Scholar]
  • 48.Pruss H, et al. : Non-resolving aspects of acute inflammation after spinal cord injury (SCI): indices and resolution plateau. Brain Pathol 2011, 21:652–660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Zrzavy T, et al. : Acute and non-resolving inflammation associate with oxidative injury after human spinal cord injury. Brain 2021, 144:144–161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Nash MS, et al. : Identification and management of cardiometabolic risk after spinal cord injury: clinical practice guideline for health care providers. Top Spinal Cord Inj Rehabil 2018, 24:379–423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Nash MS, et al. : Cardiometabolic syndrome in people with spinal cord injury/disease: guideline-derived and nonguideline risk components in a pooled sample. Arch Phys Med Rehabil 2016, 97:1696–1705. [DOI] [PubMed] [Google Scholar]
  • 52.Mirhafez SR, et al. : Serum high-sensitivity C-reactive protein as a biomarker in patients with metabolic syndrome: evidence-based study with 7284 subjects. Eur J Clin Nutr 2016, 70:1298–1304. [DOI] [PubMed] [Google Scholar]
  • 53.Pearson TA, et al. : Markers of inflammation and cardiovascular disease: application to clinical and public health practice: a statement for healthcare professionals from the Centers for Disease Control and Prevention and the American Heart Association. Circulation 2003, 107:499–511. [DOI] [PubMed] [Google Scholar]
  • 54.Frost F, et al. : Inflammatory C-reactive protein and cytokine levels in asymptomatic people with chronic spinal cord injury. Arch Phys Med Rehabil 2005, 86:312–317. [DOI] [PubMed] [Google Scholar]
  • 55.Lee MY, et al. : C-reactive protein, metabolic syndrome, and insulin resistance in individuals with spinal cord injury. J Spinal Cord Med 2005, 28:20–25. [DOI] [PubMed] [Google Scholar]
  • 56.Manns PJ, McCubbin JA, Williams DP: Fitness, inflammation, and the metabolic syndrome in men with paraplegia. Arch Phys Med Rehabil 2005, 86:1176–1181. [DOI] [PubMed] [Google Scholar]
  • 57.Morse LR, et al. : Association between mobility mode and C-reactive protein levels in men with chronic spinal cord injury. Arch Phys Med Rehabil 2008, 89:726–731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Wang TD, et al. : Circulating levels of markers of inflammation and endothelial activation are increased in men with chronic spinal cord injury. J Formos Med Assoc 2007, 106:919–928. [DOI] [PubMed] [Google Scholar]
  • 59.Nash MS, G S, Mendez AJ, Hamm LF, Lewis JE, Groah SL: Elevated CRP and vascular disease after SCI: inflammatory epiphenomenon or pathologic agent? J Spinal Cord Med 2006, 29. [Google Scholar]
  • 60.Ridker PM, Bassuk SS, Toth PP: C-reactive protein and risk of cardiovascular disease: evidence and clinical application. Curr Atherosclerosis Rep 2003, 5:341–349. [DOI] [PubMed] [Google Scholar]
  • 61.Lagrand WK, et al. : C-reactive protein as a cardiovascular risk factor: more than an epiphenomenon? Circulation 1999, 100:96–102. [DOI] [PubMed] [Google Scholar]
  • 62.Hayes KC, et al. : Elevated serum titers of proinflammatory cytokines and CNS autoantibodies in patients with chronic spinal cord injury. J Neurotrauma 2002, 19:753–761. [DOI] [PubMed] [Google Scholar]
  • 63.Davies AL, Hayes KC, Dekaban GA: Clinical correlates of elevated serum concentrations of cytokines and autoantibodies in patients with spinal cord injury. Arch Phys Med Rehabil 2007, 88:1384–1393. [DOI] [PubMed] [Google Scholar]
  • 64.Segal JL, et al. : Circulating levels of IL-2R, ICAM-1, and IL-6 in spinal cord injuries. Arch Phys Med Rehabil 1997, 78:44–47. [DOI] [PubMed] [Google Scholar]
  • 65.Goldstein RL, et al. : Clinical factors associated with C-reactive protein in chronic spinal cord injury. Spinal Cord 2017, 55:1088–1095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Tahir A, et al. : An evaluation of lipid profile and pro-inflammatory cytokines as determinants of cardiovascular disease in those with diabetes: a study on a Mexican American cohort. Sci Rep 2021, 11:2435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Lemieux I, et al. : Elevated C-reactive protein: another component of the atherothrombotic profile of abdominal obesity. Arterioscler Thromb Vasc Biol 2001, 21:961–967. [DOI] [PubMed] [Google Scholar]
  • 68.Bautista LE, et al. : Independent association between inflammatory markers (C-reactive protein, interleukin-6, and TNF-alpha) and essential hypertension. J Hum Hypertens 2005, 19:149–154. [DOI] [PubMed] [Google Scholar]
  • 69.Savoia C, Schiffrin EL: Inflammation in hypertension. Curr Opin Nephrol Hypertens 2006, 15:152–158. [DOI] [PubMed] [Google Scholar]
  • 70.Bauman WA, et al. : Coronary artery disease: metabolic risk factors and latent disease in individuals with paraplegia. Mt Sinai J Med 1992, 59:163–168. [PubMed] [Google Scholar]
  • 71.Groah SL, et al. : Cardiometabolic risk in community-dwelling persons with chronic spinal cord injury. J Cardiopulm Rehabil Prev 2011, 31:73–80. [DOI] [PubMed] [Google Scholar]
  • 72.Duckworth WC, et al. : Glucose intolerance due to insulin resistance in patients with spinal cord injuries. Diabetes 1980, 29:906–910. [DOI] [PubMed] [Google Scholar]
  • 73.DeVivo MJ, Krause JS, Lammertse DP: Recent trends in mortality and causes of death among persons with spinal cord injury. Arch Phys Med Rehabil 1999, 80:1411–1419. [DOI] [PubMed] [Google Scholar]
  • 74.Garshick E, et al. : A prospective assessment of mortality in chronic spinal cord injury. Spinal Cord 2005, 43:408–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Myers J, Lee M, Kiratli J: Cardiovascular disease in spinal cord injury: an overview of prevalence, risk, evaluation, and management. Am J Phys Med Rehabil 2007, 86:142–152. [DOI] [PubMed] [Google Scholar]
  • 76.Allen KJ, Leslie SW: Autonomic dysreflexia. In StatPearls. Treasure Island (FL); 2022. [PubMed] [Google Scholar]
  • 77.Black PH, Garbutt LD: Stress, inflammation and cardiovascular disease. J Psychosom Res 2002, 52:1–23. [DOI] [PubMed] [Google Scholar]
  • 78.Nash MS, Cowan RE, Kressler J: Evidence-based and heuristic approaches for customization of care in cardiometabolic syndrome after spinal cord injury. J Spinal Cord Med 2012, 35:278–292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Garshick E, et al. : Systemic inflammation and reduced pulmonary function in chronic spinal cord injury. Pharm Manag PM R 2011, 3:433–439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Hart JE, et al. : Cross-sectional associations of pulmonary function with systemic inflammation and oxidative stress in individuals with chronic spinal cord injury. J Spinal Cord Med 2016, 39:344–352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Savic G, et al. : Causes of death after traumatic spinal cord injury-a 70-year British study. Spinal Cord 2017, 55:891–897. [DOI] [PubMed] [Google Scholar]
  • 82.Watt JW, et al. : Survival after short- or long-term ventilation after acute spinal cord injury: a single-centre 25-year retrospective study. Spinal Cord 2011, 49:404–410. [DOI] [PubMed] [Google Scholar]
  • 83.Shavelle RM, et al. : Long-term survival of persons ventilator dependent after spinal cord injury. J Spinal Cord Med 2006, 29:511–519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Cosman BC, Stone JM, Perkash I: Gastrointestinal complications of chronic spinal cord injury. J Am ParaplegiaSoc 1991, 14:175–181. [DOI] [PubMed] [Google Scholar]
  • 85.Ebert E: Gastrointestinal involvement in spinal cord injury: a clinical perspective. J Gastrointestin Liver Dis 2012, 21:75–82. [PubMed] [Google Scholar]
  • 86.Gore RM, Mintzer RA, Calenoff L: Gastrointestinal complications of spinal cord injury. Spine 1981, 6:538–544. [DOI] [PubMed] [Google Scholar]
  • 87.Cani PD, et al. : Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 2007, 56:1761–1772. [DOI] [PubMed] [Google Scholar]
  • 88.Neves AL, et al. : Metabolic endotoxemia: a molecular link between obesity and cardiovascular risk. J Mol Endocrinol 2013, 51:R51–R64. [DOI] [PubMed] [Google Scholar]
  • 89.Puddu A, et al. : Evidence for the gut microbiota short-chain fatty acids as key pathophysiological molecules improving diabetes. Mediat Inflamm 2014, 2014, 162021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Gungor B, et al. : Intestinal microbiota in patients with spinal cord injury. PLoS One 2016, 11:e0145878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Prio TK, et al. : Asymptomatic bacteriuria in elderly humans is associated with increased levels of circulating TNF receptors and elevated numbers of neutrophils. Exp Gerontol 2002, 37:693–699. [DOI] [PubMed] [Google Scholar]
  • 92.Fouts DE, et al. : Integrated next-generation sequencing of 16S rDNA and metaproteomics differentiate the healthy urine microbiome from asymptomatic bacteriuria in neuropathic bladder associated with spinal cord injury. J Transl Med 2012, 10:174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Valentine RJ, et al. : Sex differences in the relationship between obesity, C-reactive protein, physical activity, depression, sleep quality and fatigue in older adults. Brain Behav Immun 2009, 23:643–648. [DOI] [PubMed] [Google Scholar]
  • 94.Valentine RJ, et al. : Stronger relationship between central adiposity and C-reactive protein in older women than men. Menopause 2009, 16:84–89. [DOI] [PubMed] [Google Scholar]
  • 95.Nash MS, et al. : Identification and management of cardiometabolic risk after spinal cord injury. J Spinal Cord Med 2019, 42:643–677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Bauman WA, Spungen AM: Metabolic changes in persons after spinal cord injury. Phys Med Rehabil Clin 2000, 11:109–140. [PubMed] [Google Scholar]
  • 97.Sturgeon JD, et al. : Hemostatic and inflammatory risk factors for intracerebral hemorrhage in a pooled cohort. Stroke 2008, 39:2268–2273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Gorgey AS, Gater DR: Regional and relative adiposity patterns in relation to carbohydrate and lipid metabolism in men with spinal cord injury. Appl Physiol Nutr Metabol 2011, 36:107–114. [DOI] [PubMed] [Google Scholar]
  • 99.Nash MS G DR: Exercise to reduce obesity in SCI. Top Spinal Cord Inj Rehabil 2007, 12:76–93. [Google Scholar]
  • 100.Jialal I, Devaraj S, Venugopal SK: C-reactive protein: risk marker or mediator in atherothrombosis? Hypertension 2004, 44:6–11. [DOI] [PubMed] [Google Scholar]
  • 101.Wang YH, et al. : Fasting serum levels of adiponectin, ghrelin, and leptin in men with spinal cord injury. Arch Phys Med Rehabil 2005, 86:1964–1968. [DOI] [PubMed] [Google Scholar]
  • 102.Maruyama Y, et al. : Serum leptin, abdominal obesity and the metabolic syndrome in individuals with chronic spinal cord injury. Spinal Cord 2008, 46:494–499. [DOI] [PubMed] [Google Scholar]
  • 103.Jung UJ, Choi MS: Obesity and its metabolic complications: the role of adipokines and the relationship between obesity, inflammation, insulin resistance, dyslipidemia and nonalcoholic fatty liver disease. Int J Mol Sci 2014, 15:6184–6223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Roytblat L, et al. : Raised interleukin-6 levels in obese patients. Obes Res 2000, 8:673–675. [DOI] [PubMed] [Google Scholar]
  • 105.Hansen D, et al. : Plasma adipokine and inflammatory marker concentrations are altered in obese, as opposed to non-obese, type 2 diabetes patients. Eur J Appl Physiol 2010, 109:397–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Krause JS, et al. : Allostatic load and spinal cord injury: review of existing research and preliminary data. Top Spinal Cord Inj Rehabil 2014, 20:137–146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Kern PA, et al. : Adipose tissue tumor necrosis factor and interleukin-6 expression in human obesity and insulin resistance. Am J Physiol Endocrinol Metab 2001, 280:E745–E751. [DOI] [PubMed] [Google Scholar]
  • 108.Kishore P, et al. : Adipocyte-derived factors potentiate nutrient-induced production of plasminogen activator inhibitor-1 by macrophages. Sci Transl Med 2010, 2:20ra15. [DOI] [PubMed] [Google Scholar]
  • 109.Ekstrom M, et al. : Stimulated in vivo synthesis of plasminogen activator inhibitor-1 in human adipose tissue. Thromb Haemostasis 2012, 108:485–492. [DOI] [PubMed] [Google Scholar]
  • 110.Belalcazar LM, et al. : Metabolic factors, adipose tissue, and plasminogen activator inhibitor-1 levels in type 2 diabetes: findings from the look AHEAD study. Arterioscler Thromb Vasc Biol 2011, 31:1689–1695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Alessi MC, Juhan-Vague I: PAI-1 and the metabolic syndrome: links, causes, and consequences. Arterioscler Thromb Vasc Biol 2006, 26:2200–2207. [DOI] [PubMed] [Google Scholar]
  • 112.Singh P, et al. : Leptin upregulates the expression of plasminogen activator inhibitor-1 in human vascular endothelial cells. Biochem Biophys Res Commun 2010, 392:47–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Garshick E, et al. : Plasma leptin and reduced FEV1 and FVC in chronic spinal cord injury. Pharm Manag PM R 2018, 10:276–285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Elenkov IJ, et al. : The sympathetic nerve–an integrative interface between two supersystems: the brain and the immune system. Pharmacol Rev 2000, 52:595–638. [PubMed] [Google Scholar]
  • 115.Cruse JM, et al. : Neuroendocrine-immune interactions associated with loss and restoration of immune system function in spinal cord injury and stroke patients. Immunol Res 1992, 11:104–116. [DOI] [PubMed] [Google Scholar]
  • 116.Pavlov VA, Tracey KJ: Neural regulation of immunity: molecular mechanisms and clinical translation. Nat Neurosci 2017, 20:156–166. [DOI] [PubMed] [Google Scholar]
  • 117.Meisel C, et al. : Central nervous system injury-induced immune deficiency syndrome. Nat Rev Neurosci 2005, 6:775–786. [DOI] [PubMed] [Google Scholar]
  • 118.Campagnolo DI, et al. : Altered innate immunity following spinal cord injury. Spinal Cord 2008, 46:477–481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Held KS, et al. : Impaired immune responses following spinal cord injury lead to reduced ability to control viral infection. Exp Neurol 2010, 226:242–253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Lucin KM, et al. : Impaired antibody synthesis after spinal cord injury is level dependent and is due to sympathetic nervous system dysregulation. Exp Neurol 2007, 207:75–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Lacy P, Stow JL: Cytokine release from innate immune cells: association with diverse membrane trafficking pathways. Blood 2011, 118:9–18. [DOI] [PubMed] [Google Scholar]
  • 122.Cruse JM, et al. : Facilitation of immune function, healing of pressure ulcers, and nutritional status in spinal cord injury patients. Exp Mol Pathol 2000, 68:38–54. [DOI] [PubMed] [Google Scholar]
  • 123.Street JT, et al. : Incidence of acute care adverse events and long-term health-related quality of life in patients with TSCI. Spine J 2015, 15:923–932. [DOI] [PubMed] [Google Scholar]
  • 124.Jackson AB, Groomes TE: Incidence of respiratory complications following spinal cord injury. Arch Phys Med Rehabil 1994, 75:270–275. [DOI] [PubMed] [Google Scholar]
  • 125.Cardozo CP: Respiratory complications of spinal cord injury. J Spinal Cord Med 2007, 30:307–308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Smith BM, et al. : Acute respiratory tract infection visits of veterans with spinal cord injuries and disorders: rates, trends, and risk factors. J Spinal Cord Med 2007, 30:355–361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Rh I: Near term focus. Available from: http.//www.rickhanseninstitute.org/51-what-we-do/what-we-do/264-near-term-focusAccessed. [Google Scholar]
  • 128.Failli V, et al. : Functional neurological recovery after spinal cord injury is impaired in patients with infections. Brain 2012, 135:3238–3250. [DOI] [PubMed] [Google Scholar]
  • 129.Dietz V, Fouad K: Restoration of sensorimotor functions after spinal cord injury. Brain 2014, 137:654–667. [DOI] [PubMed] [Google Scholar]
  • 130.Allison DJ, et al. : Changes in nutrient intake and inflammation following an anti-inflammatory diet in spinal cord injury. J Spinal Cord Med 2019, 42:768–777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Allison DJ, Ditor DS: Targeting inflammation to influence mood following spinal cord injury: a randomized clinical trial. J Neuroinflammation 2015, 12:204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Allison DJ, et al. : The influence of chronic inflammation on peripheral motor nerve conduction following spinal cord injury: a randomized clinical trial. Top Spinal Cord Inj Rehabil 2017, 23:377–385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Allison DJ, et al. : Targeting inflammation to influence cognitive function following spinal cord injury: a randomized clinical trial. Spinal Cord 2017, 55:26–32. [DOI] [PubMed] [Google Scholar]
  • 134.Allison DJ, et al. : Targeting inflammation as a treatment modality for neuropathic pain in spinal cord injury: a randomized clinical trial. J Neuroinflammation 2016, 13:152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.da Silva Alves Eduardo, de Aquino Lemos V, da Silva Francieli Ruiz, Lira Fabio Santos, dos Santos Ronaldo Vagner Thomathieli, Rosa João Paulo Pereira, Caperuto Erico, Tufik Sergio, de Mell Marco Tulio: Low-grade Inflammation and spinal cord injury: Exercise as therapy? Mediat Inflamm 2013, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Nash MS: Exercise and immunology. Med Sci Sports Exerc 1994, 26:125–127. [DOI] [PubMed] [Google Scholar]
  • 137.Nash MS: Immune responses to nervous system decentralization and exercise in quadriplegia. Med Sci Sports Exerc 1994, 26:164–171. [DOI] [PubMed] [Google Scholar]
  • 138.Nash MS: Known and plausible modulators of depressed immune functions following spinal cord injuries. J Spinal Cord Med 2000, 23:111–120. [DOI] [PubMed] [Google Scholar]
  • 139.Nash MS: Exercise as a health-promoting activity following spinal cord injury. J Neurol Phys Ther 2005, 29:87–103. 106. [DOI] [PubMed] [Google Scholar]
  • 140.Koury JC, et al. : Time of physical exercise practice after injury in cervical spinal cord-injured men is related to the increase in insulin sensitivity. Spinal Cord 2013, 51:116–119. [DOI] [PubMed] [Google Scholar]
  • 141.Neefkes-Zonneveld CR, et al. : Effect of long-term physical activity and acute exercise on markers of systemic inflammation in persons with chronic spinal cord injury: a systematic review. Arch Phys Med Rehabil 2015, 96:30–42. [DOI] [PubMed] [Google Scholar]
  • 142.van der Scheer JW, et al. : Effects of exercise on fitness and health of adults with spinal cord injury: a systematic review. Neurology 2017, 89:736–745. [DOI] [PubMed] [Google Scholar]
  • 143.Rosety-Rodriguez M, et al. : Low-grade systemic inflammation and leptin levels were improved by arm cranking exercise in adults with chronic spinal cord injury. Arch Phys Med Rehabil 2014, 95:297–302. [DOI] [PubMed] [Google Scholar]
  • 144.Bakkum AJ, et al. : Effects of hybrid cycle and handcycle exercise on cardiovascular disease risk factors in people with spinal cord injury: a randomized controlled trial. J Rehabil Med 2015, 47:523–530. [DOI] [PubMed] [Google Scholar]
  • 145.Bigford GE, et al. : A lifestyle intervention program for successfully addressing major cardiometabolic risks in persons with SCI: a three-subject case series. Spinal Cord Ser Cases 2017, 3, 17007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Kressler J, et al. : Understanding therapeutic benefits of overground bionic ambulation: exploratory case series in persons with chronic, complete spinal cord injury. Arch Phys Med Rehabil 2014, 95:1878–1887. e4. [DOI] [PubMed] [Google Scholar]
  • 147.Palermo AE, et al. : Clinician-focused overview of bionic exoskeleton use after spinal cord injury. Top Spinal Cord Inj Rehabil 2017, 23:234–244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Asselin P, et al. : Heart rate and oxygen demand of powered exoskeleton-assisted walking in persons with paraplegia. J Rehabil Res Dev 2015, 52:147–158. [DOI] [PubMed] [Google Scholar]
  • 149.Yang A, et al. : Assessment of in-hospital walking velocity and level of assistance in a powered exoskeleton in persons with spinal cord injury. Top Spinal Cord Inj Rehabil 2015, 21:100–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Nightingale TE, et al. : The influence of a home-based exercise intervention on human health indices in individuals with chronic spinal cord injury (HOMEX-SCI): study protocol for a randomised controlled trial. Trials 2016, 17:284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Nightingale TE, et al. : Home-based exercise enhances health-related quality of life in persons with spinal cord injury: a randomized controlled trial. Arch Phys Med Rehabil 2018, 99:1998–2006. e1. [DOI] [PubMed] [Google Scholar]
  • 152.Dinarello CA, Simon A, van der Meer JW: Treating inflammation by blocking interleukin-1 in a broad spectrum of diseases. Nat Rev Drug Discov 2012, 11:633–652. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Kopf M, Bachmann MF, Marsland BJ: Averting inflammation by targeting the cytokine environment. Nat Rev Drug Discov 2010, 9:703–718. [DOI] [PubMed] [Google Scholar]
  • 154.Bigford GE, et al. : Cardiometabolic risks and atherosclerotic disease in ApoE knockout mice: effect of spinal cord injury and Salsalate anti-inflammatory pharmacotherapy. PLoS One 2021, 16:e0246601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Arnett DK, et al. : 2019 ACC/AHA guideline on the primary prevention of cardiovascular disease: a report of the American college of cardiology/American heart association task force on clinical practice guidelines. J Am Coll Cardiol 2019, 74:e177–e232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Edwards LA, Bugaresti JM, Buchholz AC: Visceral adipose tissue and the ratio of visceral to subcutaneous adipose tissue are greater in adults with than in those without spinal cord injury, despite matching waist circumferences. Am J Clin Nutr 2008, 87:600–607. [DOI] [PubMed] [Google Scholar]
  • 157.Gorgey AS, Mather KJ, Gater DR: Central adiposity associations to carbohydrate and lipid metabolism in individuals with complete motor spinal cord injury. Metabolism 2011, 60:843–851. [DOI] [PubMed] [Google Scholar]
  • 158.Agostini L, et al. : NALP3 forms an IL-1beta-processing inflammasome with increased activity in Muckle-Wells autoinflammatory disorder. Immunity 2004, 20:319–325. [DOI] [PubMed] [Google Scholar]
  • 159.Goossens GH, et al. : Expression of NLRP3 inflammasome and T cell population markers in adipose tissue are associated with insulin resistance and impaired glucose metabolism in humans. Mol Immunol 2012, 50:142–149. [DOI] [PubMed] [Google Scholar]
  • 160.Wen H, et al. : Fatty acid-induced NLRP3-ASC inflammasome activation interferes with insulin signaling. Nat Immunol 2011, 12:408–415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Vandanmagsar B, et al. : The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nat Med 2011, 17:179–188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Abulafia DP, et al. : Inhibition of the inflammasome complex reduces the inflammatory response after thromboembolic stroke in mice. J Cerebr Blood Flow Metabol 2009, 29:534–544. [DOI] [PubMed] [Google Scholar]
  • 163.de Rivero Vaccari JP, et al. : A molecular platform in neurons regulates inflammation after spinal cord injury. J Neurosci 2008, 28:3404–3414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.de Rivero Vaccari JP, et al. : Therapeutic neutralization of the NLRP1 inflammasome reduces the innate immune response and improves histopathology after traumatic brain injury. J Cerebr Blood Flow Metabol 2009, 29:1251–1261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Kerr NA, et al. : Traumatic brain injury-induced acute lung injury: evidence for activation and inhibition of a neural-respiratory-inflammasome Axis. J Neurotrauma 2018, 35:2067–2076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Wang L, Hauenstein AV: The NLRP3 inflammasome: mechanism of action, role in disease and therapies. Mol Aspect Med 2020, 76, 100889. [DOI] [PubMed] [Google Scholar]
  • 167.Bauman WA, Spungen AM: Disorders of carbohydrate and lipid metabolism in veterans with paraplegia or quadriplegia: a model of premature aging. Metabolism 1994, 43:749–756. [DOI] [PubMed] [Google Scholar]
  • 168.Charlifue S, Jha A, Lammertse D: Aging with spinal cord injury. Phys Med Rehabil Clin 2010, 21:383–402. [DOI] [PubMed] [Google Scholar]
  • 169.Groah SL, et al. : Spinal cord injury and aging: challenges and recommendations for future research. Am J Phys Med Rehabil 2012, 91:80–93. [DOI] [PubMed] [Google Scholar]
  • 170.Wahman K, et al. : Cardiovascular disease risk and the need for prevention after paraplegia determined by conventional multifactorial risk models: the Stockholm spinal cord injury study. J Rehabil Med 2011, 43:237–242. [DOI] [PubMed] [Google Scholar]

RESOURCES