Next Article in Journal
Complex Interplay of Genes Underlies Invasiveness in Fibrosarcoma Progression Model
Previous Article in Journal
Common Mental Disorders, Functional Limitation and Diet Quality Trends and Related Factors among COPD Patients in Spain, 2006–2017: Evidence from Spanish National Health Surveys
Previous Article in Special Issue
Pathophysiology and Treatment Strategies of Acute Myopathy and Muscle Wasting after Sepsis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Chronic Critical Illness and PICS Nutritional Strategies

by
Martin D. Rosenthal
*,
Erin L. Vanzant
and
Frederick A. Moore
Division of Acute Care Surgery and the Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, FL 32610, USA
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2021, 10(11), 2294; https://doi.org/10.3390/jcm10112294
Submission received: 23 March 2021 / Revised: 14 May 2021 / Accepted: 17 May 2021 / Published: 25 May 2021
(This article belongs to the Special Issue Management of Chronic Critical Illness after Sepsis)

Abstract

:
The nutritional hallmark of chronic critical illness (CCI) after sepsis is persistent inflammation, immunosuppression, and catabolism syndrome (PICS), which results in global resistance to the anabolic effect of nutritional supplements. This ultimately leaves these patients in a downward phenotypic spiral characterized by cachexia with profound weakness, decreased capacity for rehabilitation, and immunosuppression with the propensity for sepsis recidivism. The persistent catabolism is driven by a pathologic low-grade inflammation with the inability to return to homeostasis and by ongoing increased energy expenditure. Better critical care support systems and advances in technology have led to increased intensive care unit (ICU) survival, but CCI due to PICS with poor long-term outcomes has emerged as a frequent phenotype among ICU sepsis survivors. Unfortunately, therapies to mitigate or reverse PICS-CCI are limited, and recent evidence supports that these patients fail to respond to early ICU evidence-based nutrition protocols. A lack of randomized controlled trials has limited strong recommendations for nutrition adjuncts in these patients. However, based on experience in other conditions characterized by a similar phenotype, immunonutrients aimed at counteracting inflammation, immunosuppression, and catabolism may be important for improving outcomes in PICS-CCI patients. This manuscript intends to review several immunonutrients as adjunctive therapies in treating PICS-CCI.

1. Introduction

Advances in intensive care unit (ICU) technology, support systems, care bundles, and evidence-based protocols have improved ICU survival by largely preventing early deaths [1]. Girard and Raffin in 1985 first described the transition from acute critical illness to chronic critical illness (CCI) as the need for acutely ill patients to require ongoing chronic support in the ICU setting [2]. As patients require ongoing care throughout chronic phases, the ICU setting has been extended to include care provided at long-term acute care facilities (LTACs). The clinical definition of CCI is not well established and quite variable. Most early descriptions involved patients requiring prolonged mechanical ventilation and discharge to LTACs for ventilator weaning. A variety of descriptive terms have been used in these reports such as the myopathy of critical illness, neuropathy of critical illness, and most recently the post intensive care unit syndrome. Kahn and Iwashyna et al. reported using a survey of inpatient databases that 5–8% of all ICU admissions (medical and surgical) develop CCI, accounting for greater than 380,000 annual cases, 107,000 in-hospital deaths, and over USD 26 billion in health care costs [3,4]. However, this relatively small patient population consumes over 30% of ICU resources [4,5]. This growing body of literature now has been extended to include many different types of ICU patients; however, a major omission is the limited amount of information and ongoing research related to the underlying pathobiology of CCI. The University of Florida (UF) Sepsis Critical illness Research Center (SCIRC) has focused its efforts over the past decade on characterizing CCI in surgical ICU patients who share a similar response after trauma, burns, and sepsis. When this response becomes excessive and dysregulated, it causes an injurious systemic inflammatory response syndrome (SIRS, with multiple organ dysfunctions), an immunosuppressive compensatory anti-inflammatory response syndrome (CARS, with secondary infections), and catabolic stress metabolism (with loss of lean body mass) [6]. Recent studies showed that these responses occur simultaneously and that it is the failure to return to homeostasis that characterizes the CCI clinical trajectory [7,8,9]. In a 2012 review article, UF SCIRC investigators coined the term persistent inflammation, immunosuppression, and catabolism syndrome (PICS) to provide a mechanistic framework in which to study CCI in surgical ICU patients [10]. From 2014 to 2019, they enrolled 363 septic patients who they followed longitudinally for 1 year in a NIH-funded prospective study to define the epidemiology, dysregulated immunity, and long-term outcomes of new onset sepsis in surgical ICU patients [11]. Patients were categorized into three clinical trajectories: (1) early death (within 14 days), (2) rapid recovery (RAP, ICU stay < 14 days), and (3) CCI (≥14 days of ICU stay with ongoing organ dysfunction based on SOFA score). It was observed that early mortality was surprisingly low (only 4%) and that 63% of patients experienced RAP. However, one third of survivors progressed into CCI. Unfortunately, most of the CCI patients were discharged to non-home destinations with severe functional and cognitive disabilities from which they do not recover, and 40% were dead at 1 year [12,13,14]. These poor long-term outcomes after sepsis are consistent with other recent reports [15,16,17,18,19]. CCI patients tend to be discharged frail and to suffer from significant pain, dyspnea, psychological distress, fatigue, and delirium related to their protracted ICU and hospital stay [20,21,22,23,24,25]. CCI patients who survive have poor quality of life, with many suffering from depression, cognitive impairments, complex physiologic abnormalities, and chronic organ dysfunction from which they rarely recover [24,26,27,28,29,30,31,32,33].
In 2020, Rosenthal et al., published an analysis of CCI patients from the UF SCIRC sepsis database in which it was found that, despite receiving adequate macronutrients starting early in the ICU, CCI patients did not respond the way RAP patients did. Rather, they experienced a persistent acute phase response (with high C reactive protein and negative acute phase reactants, as depicted by low albumin levels) and failed to become anabolic [34]. These data provide the rationale for additional nutritional adjuncts in PICS-CCI patients. This manuscript reviews specific nutritional adjuncts that likely are beneficial for patients with chronic diseases that experience a similar phenotype of persistent inflammation, anabolic resistance, and cachexia. These are discussed in the following sections: Protein Supplements, Specific Amino Acids, Omega 3 Fatty Acids (FA) and Specialized Pro-Resolving Mediator (SPMs) Supplementation, Probiotics, and Anabolic Agents.

2. Protein Supplements

Over the past few decades, guideline recommendations for ICU protein supplementation have increased from 0.8 g/kg/day up to as high as 2 g/kg/day (2.5 g/kg/day in renal replacement therapy) in an attempt to offset catabolism and anabolic resistance [35]. The emphasis is no longer on positive caloric balance but rather on providing specific macronutrients that drive clinical outcomes for ICU patients. During periods of physiologic stress, the body mobilizes and then catabolizes large amounts of protein from muscle that functionally debilitates patients [36]. After trauma and sepsis, resting energy expenditure peaks at around 5 days but can continue for up to 12 days, losing up to 16% of total body protein [37,38,39]. Weijs et al. showed that prescribing early high protein had survival benefit in ICU patients yet overfeeding was linked to increased mortality [40,41]. Allingstrup et al. added that a higher protein prescription (>1.46 g/kg/day vs. 1.06 or 0.79 g/kg/day) and specific amino acids supplementation (either Viamin 18 or Glavamin, Fresenius Kabi, Germany) was again associated with a lower mortality [42]. Compher and Heyland et al. reported that increased protein delivery had a significant survival benefit in nutritionally high-risk patients based off the Nutrition Risk in the Critically Ill (NUTRIC) > 5 but not significantly so in nutritionally low-risk patients [43]. In 2013, Wolfe and Deutz et al. described an “anabolic response”, where higher protein supplementation suppressed endogenous protein catabolism, and that this anabolic response was dose dependent to the higher amount of protein provided [44]. All of these studies support protein as the single most important macronutrient driving clinical outcomes in ICU patients, which likely impacts patients throughout their length of hospital stay providing benefit to PICS-CCI patients.
Adding to the rationale for higher protein supplementation in PICS-CCI was another recent publication from a Protein Summit that recommended protein prescription in the range of 1.2–2.5 g/kg/day in the ICU setting to optimize nutrition, to preserve muscle mass, and to decrease mortality [45]. Finally, Rondanelli et al. identified that the combination of exercise and protein supplementation with vitamin D and an improved omega 6 to omega 3 FA ratio (either omega 3 supplementation directly or consumption of fish > 4 times/week) maintained lean muscle mass in elderly patients [46]. This again has implications for PICS-CCI patients to combat catabolism by potentially feeding them with increasing doses of protein to improve long-term outcomes.

3. Specific Amino Acids

Arginine is a conditional amino acid with a wide range of bioactive impact but under states of physiologic stress and ongoing inflammation becomes depleted. Barbul and colleagues were early champions of arginine metabolism and reported its importance in wound healing and as an immunologic regulator [47,48,49,50,51,52]. Arginine serves as a substrate for nitric oxide (NO) production, causing vasodilation in tissue to enhance the delivery of oxygen and nutrients to healing wounds [51,52,53,54,55,56]. Additionally, arginine serves as an intra-cellular substrate for NO production in macrophages to improve bactericidal activity as well as improves T-cell function, proliferation, and maturation to reverse immunosuppression among ICU patients [47,48,49,55,57,58,59]. While arginine use in acute sepsis remains controversial, we have shown that the persistent expansion of myeloid-derived suppressor cells (MDSCs) characterizes PICS-CCI and is predictive of secondary infections and late mortality. MDSCs upregulate arginase-1, causing an arginine deficiency, which drives immunosuppression by affecting lymphocyte proliferation and maturation and renders the zeta-chain of the T-cell receptor (TCR) dysfunctional, thus causing T-cell incompetency [57,60,61,62,63,64,65,66]. PICS-CCI patients are typically well outside their sentinel septic event that results in inflammation and immunosuppression, thus consequently the unfounded controversy concerning arginine aggravating an acutely septic state is irrelevant. Therefore, providing this amino acid to PICS-CCI patients makes logical sense to promote recovery and healing and to potentially reverse some of the immunosuppression that this population endures.
Branched-chain amino acids (BCAAs: leucine, isoleucine, and valine) are another group of amino acids that, when supplemented, have demonstrated decreased muscle catabolism and hypertrophic muscle growth through increased protein synthesis [67]. Frank Cerra championed an idea of “septic auto-cannibalism” that occurred in multiple organ failure (MOF) patients despite standard parenteral nutrition (PN) and recommended using BCAA to combat the ongoing ICU muscle breakdown [68]. In a prospective randomized trial, Cerra reported that the use of BCAA-fortified PN in surgical patients improved visceral protein markers and nitrogen balance as well as absolute lymphocyte count, which is one of the laboratory parameters of PICS-CCI. As these BCAA formulas were expensive, clinical use waned until recent studies linked leucine stimulation of the Mammalian Target of Rapamycin (mTOR) pathway with increases in protein synthesis (anabolic) and inhibition of proteosomal protein breakdown (anti-catabolic) [40,69]. During PICS-CCI, leucine and potentially even beta-hydroxy-beta-methylbutyrate (HMB: a metabolite of leucine) may help decrease and potentially even reverse the catabolic nature of their pathophysiologic state [70]. It is well known that critically ill patients lose lean muscle mass at an accelerated rate while being bed bound, as described above [71]. Prolonged ICU stays, associated with the chronicity of PICS-CCI patients, only adds to the persistent catabolism, giving rise to ICU-acquired weakness and a PICS phenotype with poor rehab potential [72]. Leucine or HMB supplementation hopefully preserves or increases muscle mass and strength to improve patients rehab potential and to restore some semblance of baseline function once released from the ICU. Leucine works in concert with arginine to stimulate mTOR for its required anabolic, muscle hypertrophy effect [73]. Ultimately, in supplementing these two amino acids, the goal is to allow the synergistic effect of arginine and leucine to promote an anabolic recovery sooner than expected in PICS-CCI patients.
Glutamine supplementation may prove to be important as its pluripotent ability to serve as an antioxidant, in gluconeogenesis, and to enhance immune function is lost during times of great stress. Skeletal muscle generates the majority of endogenous glutamine, but during major catabolic insults, demand for glutamine becomes far greater than supply. This phenomenon results in glutamine becoming a conditional amino acid. Glutamine serves as the primary oxidative fuel for rapidly dividing tissues, such as small bowel mucosa, proliferating lymphocytes, and macrophages [74]. Glutamine also serves in intermediary metabolism, including the maintenance of acid–base status, as a precursor of urinary ammonia, and in nitrogen transfer for the biosynthesis of nucleotides, amino sugars, arginine, glutathione (an antioxidant), and glucosamine [75,76]. During periods of stress, glutamine can provide the carbon skeleton for gluconeogenesis and is the primary substrate for renal gluconeogenesis [77]. Some studies have suggested that glutamine supplementation boosts cell-mediated immunity through enhanced proliferation of lymphocytes while simultaneously suppressing systemic inflammation through cessation of pro-inflammatory cytokines [78]. These beneficial effects on immune function and inflammation are believed to contribute to the lower rates of infection and inflammatory complications in critically ill patients who receive glutamine supplementation [79]. Though PICS-CCI patients are typically removed from their inciting pathophysiologic insult, they remain immunosuppressed and catabolic and have a high rate of sepsis recidivism. By providing the aforementioned amino acids, the hope is to fortify PICS-CCI patients with a specific amino acid that provides substantial non-caloric benefit.

4. Omega 3 Fatty Acids (FA) and Specialized Pro-Resolving Mediator (SPMs) Supplementation

Lipids remain a crucial macronutrient and mainstay nutritional therapy for the critically ill and surgical populations, but controversy still exists regarding the digestion, absorption, and oxidation of lipids in hyperdynamic states [80]. While uncertainty may exist on which lipid formulation to deliver, there is no debate regarding the need to meet the essential FA and cellular oxidative requirements. The beneficial anti-inflammatory effects of omega 3 FAs (primarily eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA)) have been well documented in several chronic inflammatory diseases, including rheumatoid arthritis, Crohn’s disease, ulcerative colitis, lupus, multiple sclerosis, and asthma [81,82,83,84,85]. Recent evidence from an updated meta-analysis by Pradelli et al. suggests a similar benefit with omega 3 supplementation. This meta-analysis included close to 50 randomized control trails (most are surgical patients) and found that parenteral omega 3 FAs decreased hospital length of stay and ICU length of stay, overall infectious complications, and septic events and was financially cost saving for the duration of ICU patients [86,87,88]. It has also been reported in numerous human randomized clinical trials that the use of omega 3 FAs (EPA/DHA) can partially attenuate the hypermetabolic response to critical illness, can minimize lean muscle loss, can inhibit oxidative injury, and can improve outcomes by modulating the synthesis of pro-inflammatory and by providing anti-inflammatory mediators [80,89,90,91]. It appears that 2 g/day of EPA and DHA orally or 0.2 g/kg/day parenterally are effective at eliciting the desired anti-inflammatory actions; however, few studies specifically looked at dose to effect outcomes [92,93,94].
An advanced understanding of the bioactive nature of omega 3 FAs (EPA/DHA) has shown us that there is an even greater endogenous byproduct of lipid metabolism called Specialized Pro-Resolving Mediator (SPMs) [95]. These highly active SPMs are classified into three species called resolvins, maresins, and protectins [96]. They are found in nano- and picomolar concentrations and help to resolve inflammation [97,98]. Serhan and colleagues have shown that SPMs decrease inflammation by halting leukocyte infiltration, activation, and enhanced innate immune function of clearing debris, bacteria, and apoptotic cells [96,97]. SPMs hypothetically can be impactful in attenuating the dysfunctional inflammatory response and immunosuppression and in decreasing catabolism through the preservation of endogenous energy no longer diverted to the chronic systemic hyper-inflamed state observed in PICS-CCI.

5. Probiotics

ICU care, as advanced as it may seem, has limitations, which has generated research interests for a better understanding of what a healthy, diverse microbiome can provide a host during critical illness. Since 1907, when the Nobel Laureate Elie Metchnikoff first described the concept of probiotics, the overarching opinion has been that probiotics are safe and have a role in treating gastrointestinal diseases, but the use has not been supported by the literature [99]. Insight into how the stresses in ICU care, critical illness, and surgery all negatively impact the microbiome has led research teams to identify that these stresses give rise to virulent organisms collectively called the pathobiome [100,101,102,103,104,105,106,107]. Various strategies have been reported to reduce the burden of the pathobiome, but none seem as promising as probiotics.
Supplementing the critically ill and post-surgical patients with both pre- and probiotics has proven beneficial in many settings, but evidence lacks as to which specific species are superior to improve the dysbiosis and can they be beneficial to PICS-CCI? Despite the amounting research that has shown benefit, supplementing probiotics is still not considered the standard of care in all ICU settings. Nevertheless, the idea is that ‘bioecological control’ has blossomed in critically illness and polytrauma patients: to supply viable beneficial bacteria or a substrate that enhances these specific beneficial bacteria instead of trying to eliminate the pathogen [108,109]. Could this mean that, by fortifying the gut microbiota through probiotics, we could alter or enhance the recovery of PICS-CCI patients?
Anton et al. performed a meta-analysis to identify nutritional and pharmacologic interventions targeting chronic low-grade inflammation in older adults. In this analysis, he identified probiotics as having the largest impact on decreasing the serum biomarkers of inflammation (IL-6 and CRP). Angiotensin II receptor blockers and omega 3 fatty acids also significantly reduced these biomarkers but to a lesser extent compared to probiotics [110]. Other chronic states of inflammation similar to PICS-CCI are associated with inflammation, catabolism, and anabolic resistance, leading to frailty and poor long-term outcomes. It is this “inflammaging” described by Franceschi that contributes to age-related decline in functional status and increases in morbidity and mortality [111]. Perhaps a better understanding of how our gut microbiome cross talks with the body in health and illness will elucidate which specific probiotic can be most beneficial for PICS-CCI?

6. Anabolic Agents

Anabolic and anti-catabolic agents may be beneficial in mitigating persistent muscle breakdown, a pillar of PICS-CCI. Herndon and colleagues described numerous adjuncts to mitigate catabolism in the pediatric burn population that may be of value in the PICS-CCI population, including (a) growth hormone [112], (b) intensive insulin therapy [113,114], (c) oxandralone [115,116], (d) propranolol [117], and (e) exercise programs [118]. Growth hormone and, arguably, each of these adjunctive therapies have a net response to be “potent anabolic agent and salutary modulators of posttraumatic metabolic responses” [112]. These therapies preserve lean muscle mass, increase strength, enhance bone mineralization, and attenuate the hypermetabolic response to burn injury, leading to quicker recovery [113,114,119].

7. Conclusions

PICS-CCI is the high cost of ICU survival and the new, dominant chronic MOF phenotype in ICUs and LTACs. The underlying pathobiology contributing to this increasingly common CCI phenotype are being unraveled. We now know that evidence-based, gold standard nutritional protocols fail to have much of an effect on the PICS-CCI phenotype as this patient population continues to exhibit a cachexia-like response to our nutritional strategies. Commencing therapeutic strategies early in critical illness may mitigate progression into CCI. When PICS-CCI is established, combatting the pillars will likely require multiple modalities. Nutritional strategies such as the use of various immunonutrients are forthcoming and are one modality to hopefully alter the clinical trajectory of PICS-CCI. Among these strategies, the ones that hold the highest potential are protein concentration ≈2 g/kg/d, specific immuno-amino acids, omega 3 fatty acids, specialized pro-resolving mediators, probiotics, and anabolic agents.

Author Contributions

All authors aided in writing and editing the manuscript. F.A.M. developed the idea. M.D.R. collated the contributions to each section for this manuscript. E.L.V. helped edit the manuscript and add sections for the final draft. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

We acknowledge the Center for Sepsis and Critical Illness Award # P50 GM-111152 from the NIGMS.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Rosenthal, M.; Gabrielli, A.; Moore, F. The evolution of nutritional support in long term ICU patients: From multisystem organ failure to persistent inflammation immunosuppression catabolism syndrome. Minerva Anestesiol. 2016, 82, 84–96. [Google Scholar] [PubMed]
  2. Girard, K.; Raffin, T.A. The chronically critically ill: To save or let die? Respir. Care 1985, 30, 339–347. [Google Scholar] [PubMed]
  3. Kahn, J.M.; Le, T.; Angus, D.C.; Cox, C.E.; Hough, C.L.; White, D.B.; Yende, S.; Carson, S. The epidemiology of chronic critical illness in the United States. Crit. Care Med. 2015, 43, 282–287. [Google Scholar] [CrossRef] [PubMed]
  4. Iwashyna, T.J.; Cooke, C.R.; Wunsch, H.; Kahn, J.M. Population burden of long-term survivorship after severe sepsis in older Americans. J. Am. Geriatr. Soc. 2012, 60, 1070–1077. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Iwashyna, T.J.; Hodgson, C.L.; Pilcher, D.; Bailey, M.; van Lint, A.; Chavan, S.; Belomo, R. Timing of onset and burden of persistent critical illness in Australia and New Zealand: A retrospective, population-based, observational study. Lancet Respir. Med. 2016, 4, 566–573. [Google Scholar] [CrossRef]
  6. Moore, F.A.; Moore, E.E.; Billiar, T.R.; Vodovotz, Y.; Banerjee, A.; Moldawer, L.L. The role of NIGMS P50 sponsored team science in our understanding of multiple organ failure. J. Trauma Acute Care Surg. 2017, 83, 520–531. [Google Scholar] [CrossRef]
  7. Horn, D.L.; Bettcher, L.F.; Navarro, S.L.; Pascua, V.; Neto, F.C.; Cuschieri, J.; Raftery, D.; O′Keefe, G.E. Persistent metabolomic alterations characterize chronic critical illness after severe trauma. J. Trauma Acute Care Surg. 2021, 90, 35–45. [Google Scholar] [CrossRef]
  8. Jeschke, M.G.; Gauglitz, G.G.; Finnerty, C.C.; Kraft, R.; Mlcak, R.P.; Herndon, D.N. Survivors versus nonsurvivors postburn: Differences in inflammatory and hypermetabolic trajectories. Ann. Surg. 2014, 259, 814–823. [Google Scholar] [CrossRef] [Green Version]
  9. Vanzant, E.L.; Lopez, C.M.; Ozrazgat–Baslanti, T.; Ungaro, R.; Davis, R.; Cuenca, A.G.; Bihorac, A.; Leeuwenburgh, C.; Lanz, J.; Baker, H.V.; et al. Persistent inflammation, immunosuppression, and catabolism syndrome after severe blunt trauma. J. Trauma Acute Care Surg. 2014, 76, 21–29. [Google Scholar] [CrossRef] [Green Version]
  10. Gentile, L.F.; Cuenca, A.G.; Efron, P.A.; Ang, D.; Bihorac, A.; McKinley, B.A.; Moldawer, L.L.; Moore, F.A. Persistent inflammation and immunosuppression: A common syndrome and new horizon for surgical intensive care. J. Trauma Acute Care Surg. 2012, 72, 1491–1501. [Google Scholar] [CrossRef] [Green Version]
  11. Loftus, T.J.; Mira, J.C.; Ozrazgat-Baslanti, T.; Ghita, G.L.; Wang, Z.; Stortz, J.A.; Brumback, B.A.; Bihorac, A.; Segal, M.S.; Anton, S.D.; et al. Sepsis and Critical Illness Research Center investigators: Protocols and standard operating procedures for a prospective cohort study of sepsis in critically ill surgical patients. BMJ Open 2017, 7, e015136. [Google Scholar] [CrossRef] [PubMed]
  12. Gardner, A.K.; Ghita, G.L.; Wang, Z.; Ozrazgat-Baslanti, T.; Raymond, S.L.; Mankowski, R.T.; Brumback, B.A.; Efron, P.A.; Bihorac, A.; Moore, F.A.; et al. The Development of Chronic Critical Illness Determines Physical Function, Quality of Life, and Long-Term Survival Among Early Survivors of Sepsis in Surgical ICUs. Crit. Care Med. 2019, 47, 566–573. [Google Scholar] [CrossRef]
  13. Brakenridge, S.C.; Efron, P.A.; Cox, M.C.; Stortz, J.A.; Hawkins, R.B.; Ghita, G.; Gardner, A.; Mohr, A.; Anton, S.D.; Moldawer, L.L.; et al. Current Epidemiology of Surgical Sepsis: Discordance Between Inpatient Mortality and 1-year Outcomes. Ann. Surg. 2019, 270, 502–510. [Google Scholar] [CrossRef] [PubMed]
  14. Mankowski, R.T.; Anton, S.D.; Ghita, G.L.; Brumback, B.; Cox, M.C.; Mohr, A.M.; Leeuwenburgh, C.; Moldawer, L.L.; Efron, P.A.; Brakenridge, S.S.; et al. Older Sepsis Survivors Suffer Persistent Disability Burden and Poor Long-Term Survival. J. Am. Geriatr. Soc. 2020, 68, 1962–1969. [Google Scholar] [CrossRef] [PubMed]
  15. Cao, X.; Liao, X.L.; He, X.L.; Xie, Z.C.; Lu, M.Z.; Kang, Y. Predictors of Long-term Mortality and Health Related Quality of Life for Elderly Patients with Sepsis. Sichuan Da Xue Xue Bao Yi Xue Ban 2018, 49, 140–144. [Google Scholar] [PubMed]
  16. Honselmann, K.C.; Buthut, F.; Heuwer, B.; Karadag, S.; Sayk, F.; Kurowski, V.; Thiele, H.; Droemann, D.; Wolfrum, S. Long-term mortality and quality of life in intensive care patients treated for pneumonia and/or sepsis: Predictors of mortality and quality of life in patients with sepsis/pneumonia. J. Crit. Care 2015, 30, 721–726. [Google Scholar] [CrossRef] [PubMed]
  17. Karlsson, S.; Ruokonen, E.; Varpula, T.; Ala-Kokko, T.I.; Pettila, V.; Finnsepsis Study Group. Long-term outcome and quality-adjusted life years after severe sepsis. Crit. Care Med. 2009, 37, 1268–1274. [Google Scholar] [CrossRef]
  18. Winters, B.D.; Eberlein, M.; Leung, J.; Needham, D.M.; Pronovost, P.J.; Sevransky, J.E. Long-term mortality and quality of life in sepsis: A systematic review. Crit. Care Med. 2010, 38, 1276–1283. [Google Scholar] [CrossRef]
  19. Wong, H.R.; Reeder, R.W.; Banks, R.; Berg, R.A.; Meert, K.L.; Hall, M.W.; McQuillen, P.S.; Mourani, P.M.; Chima, R.S.; Sorenson, S.; et al. Biomarkers for Estimating Risk of Hospital Mortality and Long-Term Quality-of-Life Morbidity After Surviving Pediatric Septic Shock: A Secondary Analysis of the Life After Pediatric Sepsis Evaluation Investigation. Pediatr. Crit. Care Med. 2021, 22, 8–15. [Google Scholar] [CrossRef]
  20. Puntillo, K.A. Pain experiences of intensive care unit patients. Heart Lung 1990, 19, 526–533. [Google Scholar]
  21. Puntillo, K.A.; White, C.; Morris, A.B.; Perdue, S.T.; Stanik-Hutt, J.; Thompson, C.L.; Wild, L.R. Patients’ perceptions and responses to procedural pain: Results from Thunder Project II. Am. J. Crit Care 2001, 10, 238–251. [Google Scholar] [CrossRef] [PubMed]
  22. Desbiens, N.A.; Mueller-Rizner, N.; Connors, A.F., Jr.; Wenger, N.S.; Lynn, J. The symptom burden of seriously ill hospitalized patients. SUPPORT Investigators. Study to Understand Prognoses and Preferences for Outcome and Risks of Treatment. J. Pain Symptom Manag. 1999, 17, 248–255. [Google Scholar] [CrossRef]
  23. Desbiens, N.A.; Wu, A.W.; Broste, S.K.; Wenger, N.S.; Connors, A.F., Jr.; Lynn, J.; Yasui, Y.; Phillips, R.P.; Fulkerson, W. Pain and satisfaction with pain control in seriously ill hospitalized adults: Findings from the SUPPORT research investigations. For the SUPPORT investigators. Study to Understand Prognoses and Preferences for Outcomes and Risks of Treatment. Crit. Care Med. 1996, 24, 1953–1961. [Google Scholar] [CrossRef]
  24. Nelson, J.E.; Meier, D.E.; Litke, A.; Natale, D.A.; Siegel, R.E.; Morrison, R.S. The symptom burden of chronic critical illness. Crit. Care Med. 2004, 32, 1527–1534. [Google Scholar] [CrossRef] [PubMed]
  25. Nelson, J.E.; Meier, D.E.; Oei, E.J.; Nierman, D.M.; Senzel, R.S.; Manfredi, P.L.; Davis, S.M.; Morrison, R.S. Self-reported symptom experience of critically ill cancer patients receiving intensive care. Crit. Care Med. 2001, 29, 277–282. [Google Scholar] [CrossRef] [PubMed]
  26. Nelson, J.E.; Mercado, A.F.; Camhi, S.L.; Tandon, N.; Wallenstein, S.; August, G.I.; Morrison, R.S. Communication about chronic critical illness. Arch. Intern. Med. 2007, 167, 2509–2515. [Google Scholar] [CrossRef] [Green Version]
  27. Carson, S.S.; Bach, P.B. The epidemiology and costs of chronic critical illness. Crit. Care Clin. 2002, 18, 461–476. [Google Scholar] [CrossRef]
  28. Carson, S.S.; Cox, C.E.; Holmes, G.M.; Howard, A.; Carey, T.S. The changing epidemiology of mechanical ventilation: A population-based study. J. Intensive Care Med. 2006, 21, 173–182. [Google Scholar] [CrossRef]
  29. Carson, S.S.; Garrett, J.; Hanson, L.C.; Lanier, J.; Govert, J.; Brake, M.C.; Landucci, D.L.; Cox, C.E.; Carey, T.S. A prognostic model for one-year mortality in patients requiring prolonged mechanical ventilation. Crit. Care Med. 2008, 36, 2061–2069. [Google Scholar] [CrossRef] [Green Version]
  30. Van den Berghe, G. Neuroendocrine pathobiology of chronic critical illness. Crit. Care Clin. 2002, 18, 509–528. [Google Scholar] [CrossRef]
  31. Douglas, S.L.; Daly, B.J.; Gordon, N.; Brennan, P.F. Survival and quality of life: Short-term versus long-term ventilator patients. Crit. Care Med. 2002, 30, 2655–2662. [Google Scholar] [CrossRef] [PubMed]
  32. Daly, B.J.; Douglas, S.L.; Gordon, N.H.; Kelley, C.G.; O’Toole, E.; Montenegro, H.; Higgins, P. Composite outcomes of chronically critically ill patients 4 months after hospital discharge. Am. J. Crit. Care 2009, 18, 56–64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Carson, S.S.; Bach, P.B.; Brzozowski, L.; Leff, A. Outcomes after long-term acute care. An analysis of 133 mechanically ventilated patients. Am. J. Respir. Crit. Care Med. 1999, 159, 1568–1573. [Google Scholar] [CrossRef] [PubMed]
  34. Rosenthal, M.D.; Bala, T.; Wang, Z.; Loftus, T.; Moore, F. Chronic Critical Illness Patients Fail to Respond to Current Evidence-Based Intensive Care Nutrition Secondarily to Persistent Inflammation, Immunosuppression, and Catabolic Syndrome. JPEN J. Parenter. Enter. Nutr. 2020, 44, 1237–1249. [Google Scholar] [CrossRef]
  35. McClave, S.A.; Taylor, B.E.; Martindale, R.G.; Warren, M.M.; Johnson, D.R.; Braunschweig, C.; McCarthy, M.S.; Davanos, E.; Rice, T.W.; Cresci, G.A.; et al. Guidelines for the Provision and Assessment of Nutrition Support Therapy in the Adult Critically Ill Patient: Society of Critical Care Medicine (SCCM) and American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.). JPEN J. Parenter. Enter. Nutr. 2016, 40, 159–211. [Google Scholar] [CrossRef] [PubMed]
  36. Preiser, J.-C.; Ichai, C.; Orban, J.-C.; Groeneveld, A.B.J. Metabolic response to the stress of critical illness. Br. J. Anaesth. 2014, 113, 945–954. [Google Scholar] [CrossRef] [Green Version]
  37. Plank, L.D.; Hill, G.L. Sequential metabolic changes following induction of systemic inflammatory response in patients with severe sepsis or major blunt trauma. World J. Surg. 2000, 24, 630–638. [Google Scholar] [CrossRef]
  38. Monk, D.N.; Plank, L.D.; Franch-Arcas, G.; Finn, P.J.; Streat, S.J.; Hill, G.L. Sequential changes in the metabolic response in critically injured patients during the first 25 days after blunt trauma. Ann. Surg 1996, 223, 395–405. [Google Scholar] [CrossRef]
  39. Plank, L.D.; Connolly, A.B.; Hill, G.L. Sequential changes in the metabolic response in severely septic patients during the first 23 days after the onset of peritonitis. Ann. Surg. 1998, 228, 146–158. [Google Scholar] [CrossRef]
  40. Weijs, P.J.; Cynoberm, L.; DeLeggem, M.; Kreymann, G.; Wernerman, J.; Wolfe, R.R. Proteins and amino acids are fundamental to optimal nutrition support in critically ill patients. Crit. Care 2014, 18, 591. [Google Scholar] [CrossRef] [Green Version]
  41. Weijs, P.J.; Looijaard, W.G.; Beishuizen, A.; Girbes, A.R.; Oudemans-van Straaten, H.M. Early high protein intake is associated with low mortality and energy overfeeding with high mortality in non-septic mechanically ventilated critically ill patients. Crit. Care 2014, 18, 701. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Allingstrup, M.J.; Esmailzadeh, N.; Wilkens Knudsen, A.; Espersen, K.; Hartvig Jensen, T.; Wiis, J.; Perner, A.; Kondrup, J. Provision of protein and energy in relation to measured requirements in intensive care patients. Clin. Nutr. 2012, 31, 462–468. [Google Scholar] [CrossRef] [PubMed]
  43. Compher, C.; Chittams, J.; Sammarco, T.; Nicolo, M.; Heyland, D.K. Greater Protein and Energy Intake May Be Associated with Improved Mortality in Higher Risk Critically Ill Patients: A Multicenter, Multinational Observational Study. Crit. Care Med. 2017, 45, 156–163. [Google Scholar] [CrossRef] [PubMed]
  44. Deutz, N.E.; Wolfe, R.R. Is there a maximal anabolic response to protein intake with a meal? Clin. Nutr. 2013, 32, 309–313. [Google Scholar] [CrossRef] [Green Version]
  45. Hurt, R.T.; McClave, S.A.; Martindale, R.G.; Gautier, J.B.O.; Coss-Bu, J.A.; Dickerson, R.N.; Heyland, D.K.; Hoffer, L.J.; Moore, F.A.; Morris, C.R.; et al. Summary Points and Consensus Recommendations from the International Protein Summit. Nutr. Clin. Pract. 2017, 32, S142–S151. [Google Scholar] [CrossRef] [Green Version]
  46. Rondanelli, M.; Faliva, M.; Monteferrario, F.; Peroni, G.; Repaci, E.; Allieri, F.; Perna, S. Novel insights on nutrient management of sarcopenia in elderly. Biomed. Res. Int. 2015, 2015, 524948. [Google Scholar] [CrossRef]
  47. Barbul, A.; Rettura, G.; Levenson, S.M.; Seifter, E. Arginine: A thymotropic and wound–healing promoting agent. Surg. Forum 1977, 28, 101–103. [Google Scholar]
  48. Barbul, A.; Sisto, D.A.; Wasserkrug, H.L.; Efron, G. Arginine stimulates lymphocyte immune response in healthy human beings. Surgery 1981, 90, 244–251. [Google Scholar]
  49. Barbul, A.; Wasserkrug, H.L.; Sisto, D.A.; Seifter, E.; Rettura, G.; Levenson, S.M.; Efron, G. Thymic stimulatory actions of arginine. JPEN J. Parenter. Enter. Nutr. 1980, 4, 446–449. [Google Scholar] [CrossRef]
  50. Barbul, A. Arginine and immune function. Nutrition 1990, 6, 53–58. [Google Scholar]
  51. Barbul, A. Arginine: Biochemistry, physiology, and therapeutic implications. JPEN J. Parenter. Enter. Nutr. 1986, 10, 227–238. [Google Scholar] [CrossRef] [PubMed]
  52. Barbul, A.; Fishel, R.S.; Shimazu, S.; Wasserkrug, H.L.; Yoshimura, N.N.; Tao, R.C.; Efron, G. Intravenous hyperalimentation with high arginine levels improves wound healing and immune function. J. Surg. Res. 1985, 38, 328–334. [Google Scholar] [CrossRef]
  53. Albina, J.E.; Mills, C.D.; Barbul, A.; Thirkill, C.E.; Henry, W.L., Jr.; Mastrofrancesco, B.; Caldwell, M.D. Arginine metabolism in wounds. Am. J. Physiol. 1988, 254, E459–E467. [Google Scholar] [CrossRef]
  54. Alexander, J.W.; Supp, D.M. Role of Arginine and Omega-3 Fatty Acids in Wound Healing and Infection. Adv. Wound Care 2014, 3, 682–690. [Google Scholar] [CrossRef] [PubMed]
  55. Bansal, V.; Ochoa, J.B. Arginine availability, arginase, and the immune response. Curr. Opin. Clin. Nutr. Metab. Care 2003, 6, 223–228. [Google Scholar] [CrossRef] [PubMed]
  56. Barbul, A.; Lazarou, S.A.; Efron, D.T.; Wasserkrug, H.L.; Efron, G. Arginine enhances wound healing and lymphocyte immune responses in humans. Surgery 1990, 108, 331–336, discussion 36–7. [Google Scholar] [PubMed]
  57. Zhu, X.; Pribis, J.P.; Rodriguez, P.C.; Morris, S.M.; Vodovotz, Y.; Billiar, T.R.; Ochoa, J.B. The central role of arginine catabolism in T-cell dysfunction and increased susceptibility to infection after physical injury. Ann. Surg. 2014, 259, 171–178. [Google Scholar] [CrossRef]
  58. Daly, J.M.; Reynolds, J.; Thom, A.; Kinsley, L.; Dietrick-Gallagher, M.; Shod, J.; Ruggieri, B. Immune and metabolic effects of arginine in the surgical patient. Ann. Surg. 1988, 208, 512–523. [Google Scholar] [CrossRef]
  59. Morris, S.M., Jr. Arginine: Master and commander in innate immune responses. Sci. Signal. 2010, 3, pe27. [Google Scholar] [CrossRef]
  60. Taheri, F.; Ochoa, J.B.; Faghiri, Z.; Culotta, K.; Park, H.J.; Lan, M.S.; Zea, A.H.; Ochoa, A.C. L-Arginine regulates the expression of the T-cell receptor zeta chain (CD3zeta) in Jurkat cells. Clin. Cancer Res. 2001, 7 (Suppl. S3), S958–S965. [Google Scholar]
  61. Rodriguez, P.C.; Zea, A.H.; Culotta, K.S.; Zabaleta, J.; Ochoa, J.B.; Ochoa, A.C. Regulation of T cell receptor CD3zeta chain expression by L-arginine. J. Biol. Chem. 2002, 277, 21123–21129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Rodriguez, P.C.; Zea, A.H.; DeSalvo, J.; Culotta, K.S.; Zabaleta, J.; Quiceno, D.G.; Ochoa, J.B.; Ochoa, A.C. L-arginine consumption by macrophages modulates the expression of CD3 zeta chain in T lymphocytes. J. Immunol. 2003, 171, 1232–1239. [Google Scholar] [CrossRef] [Green Version]
  63. Zea, A.H.; Rodriguez, P.C.; Culotta, K.S.; Hernandez, C.P.; DeSalvo, J.; Ochoa, J.B.; Park, H.-J.; Zabaleta, J.; Ochoa, A.C. L-Arginine modulates CD3zeta expression and T cell function in activated human T lymphocytes. Cell Immunol. 2004, 232, 21–31. [Google Scholar] [CrossRef] [PubMed]
  64. Makarenkova, V.P.; Bansal, V.; Matta, B.M.; Perez, L.A.; Ochoa, J.B. CD11b+/Gr-1+ myeloid suppressor cells cause T cell dysfunction after traumatic stress. J. Immunol. 2006, 176, 2085–2094. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Scumpia, P.O.; Delano, M.J.; Kelly–Scumpia, K.M.; Weinstein, J.S.; Wynn, J.L.; Winfield, R.D.; Xia, C.; Chung, C.S.; Ayala, A.; Atkinson, M.A.; et al. Treatment with GITR agonistic antibody corrects adaptive immune dysfunction in sepsis. Blood 2007, 110, 3673–3681. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Popovic, P.J.; Zeh, H.J., 3rd; Ochoa, J.B. Arginine and immunity. J. Nutr. 2007, 137 (Suppl. S2), S1681–S1686. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Rosenthal, M.D.; Vanzant, E.L.; Martindale, R.G.; Moore, F.A. Evolving paradigms in the nutritional support of critically ill surgical patients. Curr. Probl. Surg. 2015, 52, 147–182. [Google Scholar] [CrossRef]
  68. Cerra, F.B.; Siegel, J.H.; Coleman, B.; Border, J.R.; McMenamy, R.R. Septic autocannibalism. A failure of exogenous nutritional support. Ann. Surg. 1980, 192, 570–580. [Google Scholar] [CrossRef]
  69. Cynober, L.; de Bandt, J.P.; Moinard, C. Leucine and citrulline: Two major regulators of protein turnover. World Rev. Nutr. Diet 2013, 105, 97–105. [Google Scholar]
  70. Holecek, M. Beta-hydroxy-beta-methylbutyrate supplementation and skeletal muscle in healthy and muscle-wasting conditions. J. Cachexia Sarcopenia Muscle 2017, 8, 529–541. [Google Scholar] [CrossRef] [PubMed]
  71. Puthucheary, Z.A.; Rawal, J.; McPhail, M.; Connolly, B.; Ratnayake, G.; Chan, P.; Hopkins, N.S.; Phadke, R.; Dew, T.; Sidhu, P.S.; et al. Acute skeletal muscle wasting in critical illness. JAMA 2013, 310, 1591–1600. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Rosenthal, M.D.; Rosenthal, C.M.; Moore, F.A.; Martindale, R.G. Persistent, Immunosuppression, Inflammation, Catabolism Syndrome and Diaphragmatic Dysfunction. Curr. Pulmonol. Rep. 2017, 6, 54–57. [Google Scholar] [CrossRef]
  73. Bar-Peled, L.; Sabatini, D.M. Regulation of mTORC1 by amino acids. Trends Cell Biol. 2014, 24, 400–406. [Google Scholar] [CrossRef] [PubMed]
  74. Curi, R.; Newsholme, P.; Procopio, J.; Lagranha, C.; Gorjao, R.; Pithon-Curi, T.C. Glutamine, gene expression, and cell function. Front. Biosci. 2007, 12, 344–357. [Google Scholar] [CrossRef] [Green Version]
  75. Sacks, G.S. Effect of glutamine-supplemented parenteral nutrition on mortality in critically ill patients. Nutr. Clin. Pract. 2011, 26, 44–47. [Google Scholar] [CrossRef]
  76. Ziegler, T.R.; Bazargan, N.; Leader, L.M.; Martindale, R.G. Glutamine and the gastrointestinal tract. Curr. Opin. Clin. Nutr. Metab. Care 2000, 3, 355–362. [Google Scholar] [CrossRef]
  77. Griffiths, R.D. Glutamine: Establishing clinical indications. Curr. Opin. Clin. Nutr. Metab. Care 1999, 2, 177–182. [Google Scholar] [CrossRef]
  78. Cetinbas, F.; Yelken, B.; Gulbas, Z. Role of glutamine administration on cellular immunity after total parenteral nutrition enriched with glutamine in patients with systemic inflammatory response syndrome. J. Crit. Care 2010, 25, 661.e1–661.e6. [Google Scholar] [CrossRef]
  79. Kim, H. Glutamine as an immunonutrient. Yonsei Med. J. 2011, 52, 892–897. [Google Scholar] [CrossRef] [Green Version]
  80. Calder, P.C.; Yaqoob, P. Understanding omega-3 polyunsaturated fatty acids. Postgrad. Med. 2009, 121, 148–157. [Google Scholar] [CrossRef]
  81. Calder, P. Limited impact of omega-3 fatty acids in patients with multiple cardiovascular risk factors. Evid. Based Med. 2014, 19, 18. [Google Scholar] [CrossRef]
  82. Weitz, D.; Weintraub, H.; Fisher, E.; Schwartzbard, A.Z. Fish oil for the treatment of cardiovascular disease. Cardiol. Rev. 2010, 18, 258–263. [Google Scholar] [CrossRef] [Green Version]
  83. Wilczynska-Kwiatek, A.; Bargiel-Matusiewicz, K.; Lapinski, L. Asthma, allergy, mood disorders, and nutrition. Eur. J. Med. Res. 2009, 14 (Suppl. S4), 248–254. [Google Scholar] [CrossRef] [Green Version]
  84. Turner, D.; Shah, P.S.; Steinhart, A.H.; Zlotkin, S.; Griffiths, A.M. Maintenance of remission in inflammatory bowel disease using omega-3 fatty acids (fish oil): A systematic review and meta-analyses. Inflamm. Bowel Dis. 2011, 17, 336–345. [Google Scholar] [CrossRef] [PubMed]
  85. Singer, P.; Shapiro, H.; Theilla, M.; Anbar, R.; Singer, J.; Cohen, J. Anti-inflammatory properties of omega-3 fatty acids in critical illness: Novel mechanisms and an integrative perspective. Intensive Care Med. 2008, 34, 1580–1592. [Google Scholar] [CrossRef] [PubMed]
  86. Pradelli, L.; Klek, S.; Mayer, K.; Alsaleh, A.J.O.; Rosenthal, M.D.; Heller, A.R.; Muscaritoli, M. Omega-3 fatty acid-containing parenteral nutrition in ICU patients: Systematic review with meta-analysis and cost-effectiveness analysis. Crit. Care 2020, 24, 634. [Google Scholar] [CrossRef] [PubMed]
  87. Pradelli, L.; Klek, S.; Mayer, K.; Alsaleh, A.J.O.; Rosenthal, M.D.; Heller, A.R.; Muscaritoli, M. Cost-Effectiveness of Parenteral Nutrition Containing Omega-3 Fatty Acids in Hospitalized Adult Patients From 5 European Countries and the US. JPEN J. Parenter. Enter. Nutr. 2020. [Google Scholar] [CrossRef] [PubMed]
  88. Pradelli, L.; Mayer, K.; Klek, S.; Alsaleh, A.J.O.; Clark, R.; Rosenthal, M.D.; Heller, A.R.; Muscaritoli, M. Omega-3 fatty acid-containing parenteral nutrition in hospitalized patients: Systematic Review With Meta-Analysis and Trial Sequential Analysis. JPEN J. Parenter. Enteral. Nutr. 2020, 44, 44–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Pittet, Y.K.; Berger, M.M.; Pluess, T.-T.; Voirol, P.; Revelly, J.-P.; Tappy, L.; Chiolero, R.L. Blunting the response to endotoxin in healthy subjects: Effects of various doses of intravenous fish oil. Intensive Care Med. 2010, 36, 289–295. [Google Scholar] [CrossRef] [PubMed]
  90. Wischmeyer, P. Nutritional pharmacology in surgery and critical care: “You must unlearn what you have learned”. Curr. Opin. Anaesthesiol. 2011, 24, 381–388. [Google Scholar] [CrossRef]
  91. Massaro, M.; Scoditti, E.; Carluccio, M.A.; Campana, M.C.; De Caterina, R. Omega-3 fatty acids, inflammation and angiogenesis: Basic mechanisms behind the cardioprotective effects of fish and fish oils. Cell Mol. Biol. 2010, 56, 59–82. [Google Scholar]
  92. Yusof, H.M.; Cawood, A.L.; Ding, R.; Williams, J.A.; Napper, F.L.; Shearman, C.P.; Grimble, R.F.; Payne, S.P.K.; Calder, P.C. Limited impact of 2 g/day omega-3 fatty acid ethyl esters (Omacor(R)) on plasma lipids and inflammatory markers in patients awaiting carotid endarterectomy. Mar. Drugs 2013, 11, 3569–3581. [Google Scholar] [CrossRef] [PubMed]
  93. Calder, P.C. Omega-3 polyunsaturated fatty acids and inflammatory processes: Nutrition or pharmacology? Br. J. Clin. Pharmacol. 2013, 75, 645–662. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Heller, A.R.; Rössler, S.; Litz, R.J.; Stehr, S.N.; Heller, S.C.; Koch, R.; Koch, T. Omega-3 fatty acids improve the diagnosis-related clinical outcome. Crit. Care Med. 2006, 34, 972–979. [Google Scholar] [CrossRef] [PubMed]
  95. Rosenthal, M.D.; Patel, J.; Staton, K.; Martindale, R.G.; Moore, F.A.; Upchurch, G.R., Jr. Can Specialized Pro-Resolving Mediators Deliver Benefit Originally Expected from Fish Oil? Curr. Gastroenterol. Rep. 2018, 20, 40. [Google Scholar] [CrossRef] [PubMed]
  96. Serhan, C.N.; Krishnamoorthy, S.; Recchiuti, A.; Chiang, N. Novel anti-inflammatory-pro-resolving mediators and their receptors. Curr. Top. Med. Chem. 2011, 11, 629–647. [Google Scholar] [CrossRef] [PubMed]
  97. Serhan, C.N. Pro-resolving lipid mediators are leads for resolution physiology. Nature 2014, 510, 92–101. [Google Scholar] [CrossRef] [Green Version]
  98. Calder, P.C. Eicosapentaenoic and docosahexaenoic acid derived specialised pro-resolving mediators: Concentrations in humans and the effects of age, sex, disease and increased omega-3 fatty acid intake. Biochimie 2020, 178, 105–123. [Google Scholar] [CrossRef]
  99. Williams, M.D.; Ha, C.Y.; Ciorba, M.A. Probiotics as therapy in gastroenterology: A study of physician opinions and recommendations. J. Clin. Gastroenterol. 2010, 44, 631–636. [Google Scholar] [CrossRef] [Green Version]
  100. Alverdy, J.; Zaborina, O.; Wu, L. The impact of stress and nutrition on bacterial-host interactions at the intestinal epithelial surface. Curr. Opin. Clin. Nutr. Metab. Care 2005, 8, 205–209. [Google Scholar] [CrossRef]
  101. Alverdy, J.C. During critical illness the gut does not pass the acid test. Crit. Care 2012, 16, 150. [Google Scholar] [CrossRef] [Green Version]
  102. Carlisle, E.M.; Poroyko, V.; Caplan, M.S.; Alverdy, J.; Morowitz, M.J.; Liu, D. Murine gut microbiota and transcriptome are diet dependent. Ann. Surg. 2013, 257, 287–294. [Google Scholar] [CrossRef] [PubMed]
  103. Long, J.; Zaborina, O.; Holbrook, C.; Zaborin, A.; Alverdy, J. Depletion of intestinal phosphate after operative injury activates the virulence of P aeruginosa causing lethal gut-derived sepsis. Surgery 2008, 144, 189–197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Zaborina, O.; Zaborin, A.; Romanowski, K.; Babrowski, T.; Alverdy, J. Host stress and virulence expression in intestinal pathogens: Development of therapeutic strategies using mice and C. elegans. Curr. Pharm. Des. 2011, 17, 1254–1260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Alverdy, J.; Holbrook, C.; Rocha, F.; Seiden, L.; Wu, R.L.; Musch, M.; Chang, E.; Ohman, D.; Suh, S. Gut-derived sepsis occurs when the right pathogen with the right virulence genes meets the right host: Evidence for in vivo virulence expression in Pseudomonas aeruginosa. Ann. Surg. 2000, 232, 480–489. [Google Scholar] [CrossRef]
  106. Babrowski, T.; Romanowski, K.; Fink, D.; Kim, M.; Gopalakrishnan, V.; Zaborina, O.; Alverdy, J.C. The intestinal environment of surgical injury transforms Pseudomonas aeruginosa into a discrete hypervirulent morphotype capable of causing lethal peritonitis. Surgery 2013, 153, 36–43. [Google Scholar] [CrossRef] [Green Version]
  107. Laughlin, R.S.; Musch, M.W.; Hollbrook, C.J.; Rocha, F.M.; Chang, E.B.; Alverdy, J.C. The key role of Pseudomonas aeruginosa PA-I lectin on experimental gut-derived sepsis. Ann. Surg. 2000, 232, 133–142. [Google Scholar] [CrossRef]
  108. Bengmark, S. Bioecologic control of inflammation and infection in critical illness. Anesthesiol. Clin. 2006, 24, 299–323. [Google Scholar] [CrossRef]
  109. Bengmark, S. The bioecological medicine is here to stay. About microflora, synbiotics, immunity and resistance against disease. Lakartidningen 2006, 103, 293–297. [Google Scholar]
  110. Custodero, C.; Mankowski, R.; Lee, S.; Chen, Z.; Wu, S.; Manini, T.; Echeverri, J.H.; Sabbà, C.; Beavers, D.; Cauley, J.; et al. Evidence-based nutritional and pharmacological interventions targeting chronic low-grade inflammation in middle-age and older adults: A systematic review and meta-analysis. Aging Res. Rev. 2018, 46, 42–59. [Google Scholar] [CrossRef]
  111. Franceschi, C.; Campisi, J. Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. J. Gerontol. A Biol. Sci. Med. Sci. 2014, 69 (Suppl. S1), S4–S9. [Google Scholar] [CrossRef] [PubMed]
  112. Hart, D.W.; Herndon, D.N.; Klein, G.; Lee, S.B.; Celis, M.; Mohan, S.; Chinkes, D.L.; Wolf, S.E. Attenuation of posttraumatic muscle catabolism and osteopenia by long-term growth hormone therapy. Ann. Surg. 2001, 233, 827–834. [Google Scholar] [CrossRef] [PubMed]
  113. Jeschke, M.G.; Kraft, R.; Emdad, F.; Kulp, G.A.; Williams, F.N.; Herndon, D.N. Glucose control in severely thermally injured pediatric patients: What glucose range should be the target? Ann. Surg. 2010, 252, 521–527. [Google Scholar] [CrossRef] [Green Version]
  114. Jeschke, M.G.; Kulp, G.A.; Kraft, R.; Finnerty, C.C.; Mlcak, R.; Lee, J.O.; Herndon, D.N. Intensive insulin therapy in severely burned pediatric patients: A prospective randomized trial. Am. J. Respir. Crit. Care Med. 2010, 182, 351–359. [Google Scholar] [CrossRef]
  115. Porro, L.J.; Herndon, D.N.; Rodriguez, N.A.; Jennings, K.; Klein, G.L.; Mlcak, R.P.; Clayton, R.; Crites, N.N.; Hays, J.; Andersen, C.R.; et al. Five-year outcomes after oxandrolone administration in severely burned children: A randomized clinical trial of safety and efficacy. J. Am. Coll. Surg. 2012, 214, 489–502. [Google Scholar] [CrossRef] [Green Version]
  116. Sheffield-Moore, M.; Urban, R.J.; Wolf, S.E.; Jiang, J.; Catlin, D.H.; Herndon, D.N.; Wolfe, R.R.; Ferrando, A.A. Short-term oxandrolone administration stimulates net muscle protein synthesis in young men. J. Clin. Endocrinol. Metab. 1999, 84, 2705–2711. [Google Scholar] [CrossRef] [PubMed]
  117. Herndon, D.N.; Hart, D.W.; Wolf, S.E.; Chinkes, D.L.; Wolfe, R.R. Reversal of catabolism by beta-blockade after severe burns. N. Engl. J. Med. 2001, 345, 1223–1229. [Google Scholar] [CrossRef] [PubMed]
  118. Suman, O.E.; Spies, R.J.; Celis, M.M.; Mlcak, R.P.; Herndon, D.N. Effects of a 12-week resistance exercise program on skeletal muscle strength in children with burn injuries. J. Appl. Physiol. 2001, 91, 1168–1175. [Google Scholar] [CrossRef] [PubMed]
  119. Jeschke, M.G.; Chinkes, D.L.; Finnerty, C.C.; Kulp, G.; Suman, O.E.; Norbury, W.B.; Branski, L.K.; Gauglitz, G.G.; Mlcak, R.P.; Herndon, D.N. Pathophysiologic response to severe burn injury. Ann. Surg. 2008, 248, 387–401. [Google Scholar] [CrossRef] [Green Version]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Rosenthal, M.D.; Vanzant, E.L.; Moore, F.A. Chronic Critical Illness and PICS Nutritional Strategies. J. Clin. Med. 2021, 10, 2294. https://doi.org/10.3390/jcm10112294

AMA Style

Rosenthal MD, Vanzant EL, Moore FA. Chronic Critical Illness and PICS Nutritional Strategies. Journal of Clinical Medicine. 2021; 10(11):2294. https://doi.org/10.3390/jcm10112294

Chicago/Turabian Style

Rosenthal, Martin D., Erin L. Vanzant, and Frederick A. Moore. 2021. "Chronic Critical Illness and PICS Nutritional Strategies" Journal of Clinical Medicine 10, no. 11: 2294. https://doi.org/10.3390/jcm10112294

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop