Skip to main content

Revolution in sepsis: a symptoms-based to a systems-based approach?

Abstract

Severe infection and sepsis are medical emergencies. High morbidity and mortality are linked to CNS dysfunction, excessive inflammation, immune compromise, coagulopathy and multiple organ dysfunction. Males appear to have a higher risk of mortality than females. Currently, there are few or no effective drug therapies to protect the brain, maintain the blood brain barrier, resolve excessive inflammation and reduce secondary injury in other vital organs. We propose a major reason for lack of progress is a consequence of the treat-as-you-go, single-nodal target approach, rather than a more integrated, systems-based approach. A new revolution is required to better understand how the body responds to an infection, identify new markers to detect its progression and discover new system-acting drugs to treat it. In this review, we present a brief history of sepsis followed by its pathophysiology from a systems’ perspective and future opportunities. We argue that targeting the body’s early immune-driven CNS-response may improve patient outcomes. If the barrage of PAMPs and DAMPs can be reduced early, we propose the multiple CNS-organ circuits (or axes) will be preserved and secondary injury will be reduced. We have been developing a systems-based, small-volume, fluid therapy comprising adenosine, lidocaine and magnesium (ALM) to treat sepsis and endotoxemia. Our early studies indicate that ALM therapy shifts the CNS from sympathetic to parasympathetic dominance, maintains cardiovascular-endothelial glycocalyx coupling, reduces inflammation, corrects coagulopathy, and maintains tissue O2 supply. Future research will investigate the potential translation to humans.

Introduction: a global perspective

At the present time there is no magic bullet or pharmacological therapy for controlling the bioburden of propagating inflammation from intra-abdominal sepsis.

Coccolini and colleagues (2023) [26]

Sepsis is recognised by the World Health Organization (WHO) as a global health priority across all countries and ages [26]. It is the most common cause of admission and death in the Intensive Care Unit (ICU) [91]. Each year, ~ 49 million are afflicted and 11 million patients die, with the majority occurring in low- and middle-income countries [142]. Nearly half, ~ 20 million cases, occur in children under 5 years of age, with ~ 2.9 million deaths [91, 142]. These global mortality numbers translate to ~ 1200 deaths per hour or one death every 3 s. The most common causes are infections of the respiratory tract (up to 50%), followed by the abdomen, bloodstream, renal system, skin and central nervous system (CNS) [177]. Males appear to have a higher risk of mortality than females [122], which may be due to females having a more robust cell-mediated immune response [144]. Sepsis continues to pose a significant threat to the senior population with their lower physiological reserves and multiple comorbidities [80].

Sepsis is a clinical syndrome that develops from a dysregulated host response to infection [68, 91]. It is characterized by a systemic inflammatory response syndrome (SIRS) comprising hyperinflammation, immunosuppression, immune paralysis and multiple organ dysfunction syndrome (MODS) [91, 177]. Septic shock is a further complication that leads to persistent hypotension, widespread tissue hypoperfusion, SIRS, MODS and an altered mental state [4, 22, 56, 91]. Modern core definitions of sepsis emphasize a more systemic pathobiology with underlying sub-phenotypes, each potentially requiring different management strategies [148]. These new sub-phenotypes were recently identified by Seymour and colleagues, who used artificial intelligence and machine learning clustering techniques of multiple data sets from over 20,000 patients [148]. The different phenotypes appear to reflect different patient responses to infection that may be associated with varying degrees of CNS hyperactivation, inflammation, immune dysregulation, cardiac depression, endothelial-glycocalyx activation, coagulopathy and organ supply–demand imbalances. Today, diagnosing and treating sepsis begins with identifying the type of infection, measuring the host response using biomarkers, such as C-reactive protein, procalcitonin and lactate, predicting the likelihood of organ dysfunction, fluid therapy and possible drainage/surgery for source control [91, 142]. After providing a brief history of sepsis, we will discuss its pathophysiology from a systems' perspective, and the challenges and opportunities for the twenty-first century.

Brief history

The medical profession will make early diagnosis, will insist on early intervention, will limit its surgical procedures to the least possible handling and trauma consistent with closure of the opening and relief of pus tension, will limit the duration of anaesthesia and the amount of the anaesthetic, will shorten the actual time of operation, will insure the continued absence of pus tension, will eliminate the sepsis already in the blood, restore the blood pressure and will inhibit absorption by position.

John Murphy (1908) [120] p872

Pioneer surgeon John Murphy (1857–1916) wrote this description on how to treat a patient with perforative peritonitis over 100 years ago. When we study the history of medicine, one is humbled by how far we have come in advancing knowledge, on one hand, and appreciate the long road ahead to improve current practices, on the other (Fig. 1). Despite flares of brilliance from ancient times to the renaissance, major strides did not occur until the mid-1800s when knowledge and practice became more evidence-based [40, 60, 158]. Louis Pasteur’s and Robert Koch’s germ theory of diseases, Rudolf Virchow’s medicine and cellular pathology and Claude Bernard’s unifying concept of the internal milieu all formed the basis of the modern era [60] (Fig. 1). During the latter half of the 19th century, emergent surgery for intra-abdominal peritonitis with drain tubes was advocated by Johann von Mikulicz; Robert Tait practiced aseptic techniques and lavage of the peritoneal cavity; Joseph Lister made great strides in perioperative infection-control, and John Murphy incorporated most in his surgical practice (quote above), including the use of 0.9% saline infusions to avoid dehydration (Fig. 1). Despite these advances, sepsis mortality remained high (> 70%) [60, 127, 158].

Fig. 1
figure 1

Brief history of infection and sepsis from the Renaissance to the present. Note the advances in the 19th century when asepsis, surgical practice and research were rapidly being developed for major diseases and trauma. The timeline provides a perspective of the changing ideas, practices, and outcomes from which the current thinking and treatments have developed (See text). CNS, central nervous system; MgSO4, magnesium sulfate

This clinical landscape changed in 1928, when Alexander Fleming discovered Penicillium, which launched the modern antimicrobial era [127] (Fig. 1). Advances in bacteriology and antibiotic use in the 1940s and 1950s resulted in a slight mortality reduction from sepsis and septic shock [60, 158]. In the 1960s, the molecular revolution led to an increased understanding of the underlying pathology of infection and clinical trials targeted blunting the immune and inflammatory responses [60]. In the 1980s, animal models of sepsis were introduced into basic research. Today, despite new molecular-based technologies, nanotechnology diagnostics, thousands of research papers and hundreds of clinical trials, unacceptably high mortality rates still remain [27, 155]. A new revolution is required to better understand how the body responds to an infection, identify new markers to detect its progression and discover new system-acting drugs to treat it.

Pathophysiology from a system’s perspective

We begin our systems analysis of sepsis with the central nervous system (CNS) as it is the hierarchical controller of whole-body homeostasis through the multiple feedback circuits (or axes) linking the O2 we breathe to mitochondrial ATP production. The term ‘systems’ refers to the whole body’s response to an infection, which includes its activation, progression and outcome [51,52,53]. A systems-acting drug is defined as one that treats the pathophysiological response from a systems’ perspective (see later).

CNS sympathetic hyperactivity: major controller of pathophysiology

Sepsis-associated encephalopathy is a diffuse brain dysfunction that occurs secondary to infection in the body without overt CNS infection.

Gofton and Young (2011) [62]

During an infectious challenge, immune cells and their inflammatory products modulate the hypothalamic–pituitary–adrenal (HPA) axis and activate sympathetic stress response via the nucleus tractus solitarius (NTS) [42, 49, 54, 153]. In sepsis, the CNS balance switches to sympathetic dominance with suppression of parasympathetic outflows [7, 96]. This promotes a systemic pro-inflammatory state because the parasympathetic system normally keeps inflammation at bay via activation of vagal cholinergic neurons and splanchnic/splenic nerves, known as the inflammatory reflex [78, 132, 170]). This shift in CNS balance also impacts on multiple brain-axes including the lung, heart and vasculature, gut microbiome, liver, spleen, kidney, lung and muscle (Fig. 2). Animal studies, for example, have shown that blockade of the brain renin–angiotensin–aldosterone system appears to prevent sympathetic hyperactivity and markedly attenuates LV dysfunction during sepsis [36]. Targeting the CNS to reduce its sympathetic discharge offers a potential target for future system-based therapies [96].

Fig. 2
figure 2

Schematic of CNS-control linked complications following a major infection and sepsis. Excessive inflammation and tissue damage can lead to CNS dysfunction, pulmonary injury, cardiovascular uncoupling, endothelial activation, tissue ischemia, microbiome composition changes, mitochondrial dysfunction, multiple organ failure, and ultimately death. Mortality rates are from Skei and colleagues [155]. Hyperinflammation, immune dysfunction, endotheliopathy, coagulopathy and multiple organ dysfunction are all under the control of the CNS. CNS, central nervous system; BBB, blood–brain barrier; NTS, nucleus tractus solitarius; HPA, hypothalamic-pituitary axis; LV, left ventricle; DAMPs, damage-associated molecular patterns; PAMPs, pathogen-associated molecular patterns

In addition to increased sympathetic discharge, sepsis affects brain function through neural afferents, hormones and signals from systemic immune cells and tissues (see below), which can enter the brain via a leaky blood–brain barrier (BBB) (Fig. 2) [124, 146]. This creates a hostile CNS environment of inflammation, oxidative stress and redox imbalance, which can activate glial cells, constrict the microcirculation and cause ischemia, hypoxia and structural nerve damage [104, 183]. In severe cases, the bombardment of injury signals can cause a condition known as sepsis-associated encephalopathy (SAE), defined as diffuse brain dysfunction secondary to sepsis with manifestations ranging from delirium to coma [62, 146, 183]. SAE occurs in up to 70% of septic shock patients, especially in the elderly, neonates, and patients with chronic illness [62]. Furthermore, SAE is a common cause of long-term neurological damage, such as anxiety, memory impairment, and consciousness disorders following severe sepsis [62]. Potential therapeutics could target the maintenance of BBB integrity to reduce the entry of immune cells and their products, and possibly reduce neuroinflammation and the incidence of SAE.

Increased CNS-sympathetic outflows may also reduce mesenteric blood flow to the gut [37, 177]. This is particularly relevant in sepsis because if the gut becomes ischemic, it can become leaky and bacteria or their active metabolic products (lipopolysaccharides, cytokines, neuropeptides, and protein messengers) can enter the mesenteric lymph or bloodstream and exacerbate the infectious load, increase immune dysfunction, heighten inflammation, worsen coagulopathy, and trigger immunosuppression and MODS [25, 37, 75, 156, 177]. The sepsis-induced derangements in microbial balance can themselves have a profound influence on immune function and cause harm to the host (Fig. 3) [89, 157]. Change in the host’s gut microbiome is bidirectionally linked to the CNS through vagal afferents, immune, and HPA axis modulation, and the CNS in turn can modulate the gut and enteric nervous system [32, 45, 101, 113, 117, 166]. More studies are urgently required to examine the gut-brain axis and microbiome compositional changes during sepsis, which may offer potential targets for future therapeutics [25].

Fig. 3
figure 3

A schematic of the source of pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) in response to an infection, sepsis or the trauma of surgery. The immune-driven inflammatory response is determined by the mix of cytokines and other neural and inflammatory mediators that determine the selection, activation, recruitment and fate of immune effector cells. Secondary injury is defined as a progressive process that begins with a pathogen or injury and leads to CNS dysfunction, excessive inflammation, immune dysfunction, coagulopathy, oxidative stress and mitochondrial energy deficit. Sepsis progresses in the setting of hyperinflammation, immune dysfunction, oxidative stress and redox imbalance. TLR, toll-like receptor; NLR, NOD-like receptor; RAGE, receptor for advanced glycation end products; CLR, C-type lectin-like receptor; RLR, RIG-I-like receptor; NK cell, natural killer cell; ILC, innate lymphoid cell; ARDS, acute respiratory distress syndrome; PIICS, Persistent Inflammation, Immunosuppression, and Catabolism Syndrome; MODS, multiple organ dysfunction syndrome

Host response to infection: immune cell activation and inflammation

Except on few occasions, the patient appears to die from the body’s response to infection rather than from it.

Sir William Osler (1904) [128]

William Osler’s statement over 100 years ago remains the cornerstone of our thinking today. It is the body’s response to infection that is the main determinant of outcome, not the bacterial challenge per se. This helps to explain why a sepsis patient, despite successful removal of infectious foci, often fails to respond to ongoing treatments [27]. The early systemic response to an infection involves recruitment of inflammatory leukocytes to phagocytose the invading bacteria and associated cellular debris. Initially, neutrophils, which reside in large numbers in the circulation, are recruited and quickly followed by bone marrow-derived monocytes and macrophages to resolve the threat and restore homeostasis [42, 139]. Other immune cells are also recruited, such as dendritic cells, natural killer (NK) cells, B-cells, T-cells and innate lymphoid cells (ILC) [6, 22], however, their involvement in sepsis is beyond the scope of the present review (Fig. 2).

If an infection, like sepsis, overwhelms the body’s defences, innate immune cells continue to infiltrate the vital organs of the body, including the brain, resulting in collateral tissue damage from cytokines, immune modulators, complement, oxidants, proteases, and toxic extracellular traps (Fig. 2) [84]. The cytokine ‘storm’ that ensues comprises a profound excess of proinflammatory over anti-inflammatory cytokines release into the circulation (Fig. 2). Key mediators include interleukin (IL-1β), tumor necrosis factor (TNF)-α, IL-6, IL-4, IL-8, IL-17, IL-18, and IL-10, [152], as well as platelet-activating factor, complement factors and reactants of coagulopathy and fibrinolysis [179]. If concomitant anti-inflammatory processes are not activated sufficiently, the cytokine storm is often followed by persistent lymphopenia and immunosuppression or paralysis, which involves metabolic reprogramming of many different cell types, immune cell depletion, cellular apoptosis and T cell exhaustion [49, 54, 110, 177]. A number of potential targets for resolution of inflammation are underway [138, 186]). Razazi and colleagues, for example, have identified in septic shock patients that activation of the IL-17/interferon (IFN) pathway and vascular endothelial growth factor (VEGF) strongly correlates with i) early sepsis resolution (reduced lactate) and ii) improved ICU survival [138]. Another target area that appears to show promise is metabolic reprogramming of immune cell populations for switching the host’s injury phenotype to a healing one [92, 102, 103].

Source of pathogen- and injury-generated signals

The two main sources of molecular signals in sepsis that activate early immune cell recruitment and drive inflammation are: 1) pathogen-associated molecular patterns (PAMPs) from the invading infectious microbes, and 2) damage-associated molecular patterns (DAMPs) from tissue injury [42, 177] (Fig. 3). PAMPs and DAMPs are evolutionary conserved motifs that make foreign pathogens or tissue damage recognizable by the host [42, 82, 112]. PAMPs include bacterial lipopolysaccharide (LPS), flagellin and lipoteichoic acid, viral RNA and DNA, surface glycoproteins, lipoproteins, and other membrane components [10, 82, 177]. DAMPs from damaged cells and tissues caused by the infection include fibrinogen, annexins, platelet components, fibronectin, S100 proteins, syndecan-1, F-actin, adenosine triphosphate (ATP), histones, deoxyribonucleic acid (DNA), mitochondrial transcription factor A (TFAM), mitochondrial reactive oxgen species (mitoROS), cytochrome C, IL-1α, high mobility group box protein 1 (HMGB1), heparan sulfate, tenascin C, defensins, amyloid-β, and many others [141]. Another source of PAMPs and DAMPs are derived from extracellular vesicles (EVs) released by pathogens, immune or endothelial cells, platelets and damaged host cells [15, 165, 174].

How do host immune cells sense danger? PAMPs and DAMPs are detected by the host’s pattern recognition receptors (PRRs) located on the surface or in the cytoplasm of immune cells [160, 177]. PRRs are the body’s “sensors” of a threat and communicate it to the host via immune cell activation. These sensors include toll-like receptors (TLRs), RIG-I-like receptors (RLRs), NOD-like receptors (NLRs), C-type lectin-like receptors (CLRs), receptors for advanced glycation end products (RAGE) and cytosolic DNA sensors (Fig. 3) [1, 86, 173]. Another important PRR that senses a wide range of PAMPs and DAMPs is the NLR family pyrin domain containing 3 (NLRP3) protein that activates a cytoplasmic multiprotein platform assembly known as the inflammasome [85], which if overexpressed can lead to the death of the patient [181]. Importantly, PAMPs and DAMPs are not mutually exclusive, and immune cells may express co-receptors and accessory molecules that form ‘partnerships’ to coordinate an immune response [134]. The extra barrage of DAMPs from an emergent laparotomy, if required, in a sepsis patient is an additional burden rarely mentioned as a potential target to improve outcomes [168] (see later). Identifying the different PAMPs and DAMPs in the blood of sepsis patients may offer a new early diagnostic treatment window for personalized care before the cytokine storm develops or tissue blood flow and O2 becomes limiting [15, 165, 174].

Endothelial-glycocalyx: a sensor and effector of immune activation

In acute inflammation, we find, as a general rule, vascular dilatation accompanied by an active condition of the endothelium of the vessel-walls and an exudation with diapedesis, that is to say, three events which concur in producing a considerable afflux of leucocytes towards the injured spot.

E. Metchnikoff (1893) [116] p171

During sepsis, immune cells and their inflammatory products activate the endothelium that result in shedding of its ‘fuzz-like’ glycocalyx, indicated by serum elevations in syndecan-1 and soluble-thrombomodulin [81, 83, 179]. This is called sepsis-induced endotheliopathy [83]. The glycocalyx is negatively charged and anchored to the single layer of endothelial cells that forms the nexus between the vasculature and the tissues [88, 176]. It covers a vast surface area of over 55,000 m2 [49, 54]. During sepsis, the activated endothelium becomes more adhesive, leaky, pro-apoptotic, pro-inflammatory, pro-thrombotic and vasoactive (Fig. 3) [81, 83, 105, 123]. Glycocalyx shedding is facilitated by stress-activated membrane-bound enzymes, called sheddases, in response to pro-inflammatory cytokines (e.g. TNF-α and IL1-β), reactive oxygen species (ROS) (e.g. superoxide, hydroxyl radical), and by aggressive fluid resuscitation [171]. Of clinical significance, it appears that glycocalyx shedding can repair itself quickly [185]. Luft states that “these cells usually are able to replace their missing coats in a matter of minutes” [106]. However, very little is known about the loss and recovery of the glycocalyx during an infection or sepsis [171].

In critically ill patients, sepsis-induced endotheliopathy is sometimes associated with vascular microthrombosis mediated by platelet activation and the endothelial release of von Willebrand factor (vWF) multimers, which in turn impairs O2 delivery to mitochondria. Excessive production of vWF multimers become anchored to endothelial cells as elongated strings and form platelet-vWF complexes known as “microthrombi” [23, 81]. If the pathology becomes more diffuse and systemic, it can lead to a lethal condition known as disseminated intravascular coagulopathy (DIC) (see below). Reducing the early activation of the endothelial glycocalyx or facilitating rapid recovery after shedding may be potential targets for new therapeutics.

Sepsis-induced coagulopathy: a dynamic entity that evolves over time

Reconstituted systems are as realistic as our insight into the clotting mechanism allows: extrapolation to physiology should therefore be regarded with due suspicion.

Hemker and colleagues [76], p171

CNS dysfunction, inflammation, endotheliopathy and coagulopathy are all functionally linked through common pathways involved in the regulation of tissue factor (TF) [23, 47, 48, 100, 178, 179]. The common pathways include the TF inhibition pathway, platelet inhibition pathway, the heparin-antithrombin III system, thrombomodulin/protein C system and fibrinolytic pathways (Fig. 4) [47, 48, 123, 126]. Inflammasome-activated pyroptotic macrophages [181], microbial agents, cytokines and complement factors can further increase TF levels, which can activate the endothelial-glycocalyx and aggravate coagulopathy [177,178,179]. Wu and colleagues further showed in their mouse sepsis model that inhibition of TF abolishes inflammasome-mediated blood clotting and protects against death [172, 181].

Fig. 4
figure 4

Coagulopathy is a systemic pathological condition in which the blood’s ability to clot is impaired with varying degrees of fibrinolysis. The schematic illustrates the different sepsis-induced phenotypes around the Thrombomodulin (TM)-thrombin switch (1) [47, 48]. The TM-thrombin “switch” regulates coagulation and fibrinolysis in both directions depending on different activators and inhibitors at the thrombin-TM active sites (EFF-like domains) [47, 48]. During an early infection, patients appear to have a procoagulable phenotype which may form from activation of Thrombin-Activatable Fibrinolysis Inhibitor (TAFI) (2), which decreases plasmin levels (3) and increases fibrinogen to form a stronger a stronger clot. As infection progresses the phenotype may change to a more hypocoagulable state where fibrinogen is decreased, D-dimers increase (fibrinolysis), and in extreme cases progresses to a specific hypocoagulation dominated by hyperfibrinolysis with microvascular fibrin deposits (DIC). The phenotypic change from a hyper- to hypo-coagulable state to disseminated intravascular coagulopathy (DIC) appears to be associated with a transition from a TF-dominated inflammatory microenvironment, favoring EGF-like Domain 3–6), to a non-TF dominated environment, favoring EGF-like Domain 4–6, with high mortality. This hypothesis requires knowledge of cytokines, immune cells, tPA, PAI-1, α2-antiplasmin, fibrinogen, TAFI levels and remains to be tested. Drugs to modulate the thrombin-TM “switch” following infection and sepsis are urgently required. TPA: tissue plasminogen activator; PAI-1: plasminogen activator inhibitor-1; WVF: Von Willebrand factor; S100A10: S100 calcium binding protein A10; FVIII: Factor VIII; EPCR: endothelial protein C receptor; FDP: FDP: fibrin degradation product

Diagnosing sepsis-induced coagulopathy, like trauma-induced coagulopathy (TIC), has undergone major developments in the last 10 years as whole blood viscoelastic methods have replaced the older unreliable plasmatic methods of prothrombin time (PT), activated partial thromboplastin time (aPTT) and international normalized ratio (INR) [14, 46,47,48]. Sepsis-induced coagulopathy is not a static state but a dynamic one with multiple phenotypes that can change over time (Fig. 4) [14, 47, 48, 100]. In the rotational thromboelastometry (ROTEM) study of Davies and colleagues, they examined 100 ICU patients (50 with sepsis, 20 with severe sepsis and 30 with septic shock) and found increased sepsis severity was associated with shift from a hypercoagulable to hypocoagulable state, with no change in maximum clot firmness [35]. In septic shock, fibrinolysis was markedly impaired towards a bleeding phase, and was significantly associated with 28-day mortality [35]. Anatomopathologic fibrin deposition was not evaluated at autopsy to assess DIC. In 2023, Bui-Thi and colleagues undertook another prospective, observational, single-center study and reported 73% of 161 patients had sepsis/septic shock [14]. ROTEM showed 26% were hypercoagulable, 55% were hypocoagulable, 14% had mixed hypo-hypercoagulation patterns, and 19% were hyperfibrinolytic [14].

The different coagulopathies appear to reflect different timings and severity of infection and sepsis. An early common phenotype in septic patients appears to be a hypercoagulable subtype characterized by prolonged initial clot time with increased maximum clot firmness (MCF) and high fibrinogen levels (Fig. 4). Bui-Thi and colleagues further showed that progression from sepsis to severe sepsis was accompanied by a shift from hyper- to hypo-coagulability with fibrinolysis [14]. The thrombelastography (TEG) study of Luo and colleagues confirmed a hypocoagulopathy in severe sepsis patients which, if present at hospital admission, was an independent risk factor for 30-day mortality [107]. Possible mechanisms responsible for the switch from a hyper- to hypo-coagulopathy are summarized in Fig. 4 [47, 48].

A common mistake in the literature is to equate hypocoagulopathy and fibrinolysis (a bleeding phenotype) with disseminated intravascular coagulopathy (DIC) [47, 48, 100, 125]. DIC is a rare and specific phenotype with diffuse anatomopathologic fibrin deposition in small and mid-size vessels (Fig. 4) [46, 74, 99, 163]. Clinically, DIC continues to be diagnosed using a scoring system involving PT, platelet count, fibrinogen, and D-dimer levels [13, 167, 172]. However, without evidence of intravascular fibrin deposition, diagnosis may not be DIC and impact on optimal patient treatment. We propose the concept of DIC should be confined to a phenotype with confirmed microvascular fibrin deposits.

Understanding coagulopathy during sepsis is in its infancy. More research in clinically-relevant animal models and high quality prospective randomized human trials are urgently required to characterize the different early and late phenotypes, and their progression to sepsis and septic shock, as well as sex differences. With more high-quality studies, there is a strong potential that the information can lead to more effective personalized and goal-directed treatments.

Multiple organ dysfunction syndrome (MODS): not a single event but a systems failure

Of those who die, most are from multiple organ failure which is a still poorly understood consequence rather than the immediate effect of infection.

Coccolini and colleagues (2023) [27]

To highlight the lethality of MODS during sepsis, the 2023 guidelines redefined sepsis as “life-threatening organ dysfunction resulting from a dysregulated host response to infection” [143]. In 1975, Arthur Bauer introduced the term MODS as multiple physiological derangements [8]. Today, MODS is considered a clinical syndrome characterized by the development of progressive and potentially reversible physiologic dysfunction in two or more organs or organ systems [19, 95]. Organ dysfunction syndromes include encephalopathy, acute respiratory distress, myocardial infarction, hepato-renal syndrome, acute necrotizing pancreatitis, acute adrenal insufficiency, rhabdomyolysis, and muscle wasting syndrome (catabolic response) [19, 95].

At the cellular level, MODS develops from persistent tissue hypoperfusion and loss of mitochondrial integrity [49, 54]. ATP is no longer fully replenished leading to ischemia, hypoxia, organ dysfunction, and possible failure. Mitochondrial dysfunction includes decreased proton pumping across the inner mitochondrial membrane, collapsed membrane potential, opening of the mitochondrial permeability transition pore, Ca2+ loading, loss of cytochrome C, release of apoptotic-cascade inducing factors, and increased DAMPs and ROS, which exacerbates immune dysfunction, inflammation and coagulopathy [49, 54, 73, 79]. Sepsis also impairs mitochondrial biogenesis and mitophagy, resulting in insufficient renewal of mitochondria, which further impacts cellular respiratory capacity and organ dysfunction. Fever, a manifestation of sepsis, is a result of uncoupling of muscle mitochondria which leads to generation of heat, not energy, and helps to explain muscle wasting despite a high caloric intake [79]. Targeting mitochondrial dysfunction in muscle, and other organs, may offer novel and valuable targets for sepsis.

Mitochondria are not only the cell’s powerhouses. In immune cells, mitochondrial activity regulates their activation, differentiation and survival [94, 102, 103, 114]. Expression of key mediators involved in regulation of mitochondrial function (Sirt1/3, Ampk, Pgc1, Nrf1, Tfam, Mtco3, Nr3c1), for example, are significantly reduced in leukocytes from septic patients [20, 102, 103]. Sepsis-induced mitochondrial dysfunction leads to metabolic reprogramming and altered functional capacity of immune cells, heightened inflammation and immunosuppression [94, 102, 103]. Therapies that target recovery of mitochondrial function may offer a novel approach to reverse leukocyte dysfunction in sepsis [3, 114].

Cardiovascular dysfunction after sepsis: the puzzle of myocardial depression

There has been a tendency to equate shock, regardless of its origin, with a low cardiac output (CO) and high total peripheral resistance (SVR). While our experience suggests that this is true of hypovolemic and cardiac shock, the same cannot be said of the septic form.

Wilson and colleagues (1965) [180]

Cardiovascular collapse is a major reason for mortality in septic shock patients (Fig. 3) [77]. In the early 1960s, two distinct hypotensive phases of septic shock were characterized; the first was a warm dry skin, tachycardic condition (“warm” shock), and the second phase was a cold clammy skin with a thready pulse with hypotension (“cold” shock) [109]. Using this classification, septic shock initially went through an early hyperdynamic phase (high cardiac output, CO) and either recovered or deteriorated into cardiovascular collapse (low CO) [137]. In 1965, Wilson and colleagues challenged this view by reporting that septic shock patients had normal or elevated CO with low systemic vascular resistance (SVR), and very rarely had a low CO [180]. This was highly controversial and contrary to hemorrhagic/cardiogenic shock, which is characterized by a low CO and high SVR [137]. Using nuclear imaging techniques and thermodilution methods, Joseph Parrillo’s group confirmed Wilson’s findings and showed that septic shock patients maintained high CO and low SVR [28, 60, 130]. The group also reported that 75% of patients had a depressed left ventricular ejection fraction (LVEF) in the first few days after the onset of septic shock [130]. Sepsis-induced myocardial depression appears to have all the hallmarks of myocardial stunning after coronary artery bypass surgery [43, 175].

Today, sepsis-induced cardiomyopathy is an acute syndrome of myocardial depression that occurs early after the onset of septic shock and normally resolves in 7–10 days [77]. It occurs in ~ 50% of septic patients and characterized by LV dilatation and depressed LVEF with maintained coronary blood flow [77, 184]. Myocardial depression in sepsis patients is also associated with higher aortic conduit stiffness [64, 87], which may be responsible for reduced venticulo-arterial (VA) coupling and reduced blood flow and O2 to the tissues [135]. VA coupling is defined as the ratio of arterial elastance (Ea) to left-ventricular elastance (Ees), and can be derived from routine echocardiography [67]. The advantage of VA coupling over LVEF or CO is that it provides arterial load properties in addition to LV function [24, 67]. New therapeutics are required to target VA coupling in septic patients, which may prevent myocardial depression and impairment of vascular reactivity [151].

There are at least five main hypotheses of myocardial depression (Table 1). While all five are plausible, they are not mutually exclusive and appear to involve alterations in Ca2+ handling, ATP replenishment and myofilament function [9, 17, 64, 70, 77, 93, 115, 131, 138, 145, 159, 161, 182]. The future challenge is finding which one or more of these mechanisms contribute to the decline in myocardial depression and VA uncoupling seen clinically.

Table 1 Five hypotheses for sepsis-induced myocardial depression in the setting of inflammation and immune dysfunction. All five are not mutually exclusive

Fluid resuscitation: doing more harm than good?

Recently, the safety of intravenous fluids in patients with sepsis has been called into question with both prospective and observational data suggesting improved outcomes with less fluid or no fluid.

Byrne and Van Haren (2017) [16]

Current supportive strategies for severe sepsis patients may include an early and goal-directed fluid resuscitation bundle, mechanical ventilation, inotropic and vasopressor therapies, blood cell transfusions, anti-fibrinolytics and mechanical ventilation, and possible renal support [91, 177]. The primary goal of fluid therapy is to reduce dehydration, restore circulating blood volume, optimize cardiovascular function and improve tissue O2 (Fig. 5) [16]. Dehydration is common in older adults, often necessitating an initial 500 mL crystalloid bolus [80]. Prior to 2001, use of fluid resuscitation was largely based on historical experience without empirical support from either animal studies or clinical trials [16]. The first supportive evidence for fluid therapy came from the human study of Rivers and colleagues that showed a 16% mortality reduction in septic shock patients [140]. Unfortunately, the survival benefit was not supported in larger independent trials, including the ProCESS, PROMISE and ARISE trials [108].

Fig. 5
figure 5

Schematic of 2016–2021 Surviving Sepsis Campaign guidelines that suggest initial resuscitation of at least 30 mL/kg of isotonic crystalloid fluid within the first 3 h of sepsis identification to restore circulating fluid volume and optimize stroke volume. However, there is a paucity of high-quality data to support this clinical practice. The significant heterogeneity of sepsis and the reports that ~ 50% are non-responders makes the recommendation highly problematic. Current evidence indicates that administration of large fluid volumes to the critically ill may cause harm by exacerbating secondary injury (see text). IV, intravenous; CNS, central nervous system; ADP, adenosine triphosphate; ATP, adenosine triphosphate

Despite the lack of clinical evidence, the 2016–2021 Surviving Sepsis Campaign guidelines continued to promote fluid therapy for septic shock patients (Fig. 5) [58]. The guidelines proposed an initial resuscitation of at least 30 mL/kg of isotonic crystalloid fluid during the first 3 h to restore circulating fluid volume and optimize stroke volume [108]. Administering an IV fluid volume up to 60% of the normal blood volume to a 100 kg septic patient over 3 h has little or no clinical support and may be associated with higher mortality [58]. The Surviving Sepsis campaign has now downgraded this recommendation from strong to weak, although the practice appears to continue in many hospitals worldwide [58].

Today, fluid resuscitation may cause harm to some patients. Contrary to its name, normal saline is not normal, and the volumes and timings are not effective in critically ill patients [11, 16, 108, 121, 149, 150]. Large aggressive fluid volumes create dilutional coagulopathy, fluid overload, and pathogenic pulmonary and tissue edema, acute kidney injury, prolonged ICU stays and higher mortality [55, 108, 149, 150]. If normal saline was evaluated today by the European, USA and Australian regulatory bodies, there is a high chance it was not be approved for human use. The harmful effects of normal saline solutions were recognized over 100 years ago by George Evans when he wrote in 1911:One cannot fail to be impressed with the danger of such procedure, if one observes the utter recklessness with which salt solution is frequently prescribed, particularly in the postoperative period, without previous knowledge of the condition of blood pressure, the ability of the heart to handle large amounts of fluid successfully, or the functional capacity of the kidneys to excrete the large amount of chloride thus formed on them” [5, 57]. More recently, vasopressor agents that increase cardiac contractility or vasoconstriction have also come under clinical scrutiny that include increased cardiac workload, arrhythmias and vasoconstriction-related tissue ischemia [149, 150].

Another vexing problem with the ongoing use of fluid therapies is that up to 50% of patients are non-responders (Fig. 5), meaning they fail to increase preload and stroke volume with IV fluid infusions, and therefore fails to improve tissue O2 supply [72, 111]. Unfortunately, there have been few high-quality clinical trials comparing non-responders and responders [55]. Some studies recommend the use of a lung ultrasound, echocardiographic assessment, urine output, and other measures, to prevent fluid overload in the ICU in an attempt to improve patient stratification and optimize treaetment [55].

Trauma of surgery: a forgotten confounder of poor patient outcomes

It should be remembered always that the patient who has been in shock and resuscitated, and then operated upon, is in a precarious state. His nervous system has been disturbed not only by the original trauma, but also by the low nutrient flow of blood, and by the surgical procedures incidental to operation (our italics).

Walter B. Cannon (1923) Quoted from Traumatic Shock [18] p192

Patients with severe abdominal sepsis often require an emergency laparotomy, which is associated with higher mortality and morbidity compared to less invasive procedures (Fig. 2) [12, 59, 69, 90, 136, 154, 162]. Possible reasons for poorer outcomes include the trauma of surgery itself amplifying immune cell activation, increasing inflammation, coagulopathy and MODS [39, 41]. The extra barrage of DAMPs comes from tissue damage from the first incision, organ manipulation, and surgical correction and drain (Fig. 2) [2]. Torp and colleagues have recently discussed how tissue injury from surgery triggers a generalized inflammatory response and the role mitochondrial DAMPs (mDAMPs) play in worsening its pathophysiology [168]. In addition, surgical site infections occur in up to 35% of patients, which further complicates recovery [2]. Another aspect of the trauma of surgery that is rarely discussed is that the anesthetized brain is still “awake” to the circulating DAMPs from the surgery itself [30, 41], which may pass through the leaky BBB and activate CNS dysfunction and lead to altered mental states [4, 56, 91]. The trauma of surgery aggravates an already precarious state, which is magnified further in older patients with multiple comorbidities [41]. This is an area that requires new therapies to reduce the host’s stress response of surgery [31, 38, 41, 61, 71, 133, 164].

Future consideration for drug development: from symptoms to system

Clinical study of antiinflammation strategies to treat sepsis has been characterized by a predictable cycle of abundant clinical failures punctuated by an intermittent positive result.

Shapiro and colleagues (2023) [149, 150]

Notwithstanding the ongoing challenges in treating sepsis, great strides are being made in personalized care based on blood biomarkers [143]. Individualized treatments have also progressed in the chronic, immunosuppressive stage of sepsis responsible for later-stage morbidity and mortality [143]. Notwithstanding these advances, there remains few safe and effective drugs for the early treatment of sepsis [92, 102, 103, 149, 149, 150, 150]. For example, there are only a handful of drugs to reduce excessive inflammation and immune dysfunction. Non-steroidal anti-inflammatory drugs (NSAIDs), COX-2 inhibitors and TNF-α inhibitors do not appear to be pro-resolving, and may in fact exacerbate the proinflammatory process [129]. A relative new area of drug design is targeting metabolic reprogramming of immune cells responsible for hyperinflammation and immunosuppression [102, 103]. The future challenge is to safely and effectively translate these immunometabolism-altering drugs to improve patient outcomes in the hospital setting.

Why have there been so few advances in pre-clinical drug development for sepsis and translation to humans? Three possible reasons include: 1) the widespread use of specific-pathogen free (SPF) animal models with their altered microbiomes and immature immune systems that do not represent the human condition [45, 47, 48, 51,52,53], Conventionally bred and housed animals should be used if human translation is the end-game [51,52,53], 2) the flawed practice of the treat-as-you go mindset and single nodal drug targeting, which ignores the complexity of the system [49,50,51,52,53,54], and 3) poor clinical trial design that does not represent the heterogeneity of patient responses to sepsis [21, 143]. With respect to single-nodal targets, we will give one example. It is well established that the IL-1 receptor is a key amplifier of inflammation [149, 150], and its inhibition may resolve the cytokine storm. A drug that inhibits the IL-1-receptor, anakinra, has an excellent safety record in humans, however, it has failed to show a survival benefit after sepsis or COVID-19 [149, 150]. The ‘single-step’ drug target approach can be traced back to the molecular revolution of the 20th century, which began in around 1953 after the discovery of DNA [51,52,53]. Nobel Laureate Sir Francis Crick embodied this highly mechanistic mindset when he wrote “the ultimate aim of the modern movement in biology is to explain all biology in terms of physics and chemistry” [29]. From a molecular standpoint Crick was correct, however, its relevance to the workings of the whole body has not kept pace [51,52,53]. The advent of the “Omic” technologies to drill deeper into molecular mechanisms has occurred at the expense of systems analysis. Reductionism is important in breaking a complex system into its simpler parts, but it does not do away with the system. New systems-based therapies are urgently required to treat sepsis.

What would a systems-based drug look like? Ideally, a systems-based drug would blunt the CNS-linked feedback circuits (or axes) that drive secondary injury and poor patient outcomes. The drug would reduce the CNS stress response, maintain BBB integrity, promote CNS-cardiovascular coupling, prevent myocardial depression, protect the endothelial-glycocalyx, reduce excessive inflammation, reduce immune dysfunction, correct coagulopathy, and deliver sufficient O2 to tissue mitochondria [49, 50, 54]. The drug might also involve reprogramming immune cell metabolism and placing a ‘brake’ on the hyperinflammatory response and permitting anti-inflammatory processes to resolve the host’s responses to sepsis. No such systems-acting drug exists.

We have been developing an adenosine, lidocaine and magnesium (ALM) fluid therapy for hemorrhagic shock, traumatic brain injury (TBI) [44, 50,51,52,53], burns [34, 51,52,53], orthopedic trauma [118, 119] and the trauma of surgery [33]. We have shown the drug shifts sympathetic hyperactivity to parasympathetic dominance in the rat model of non-compressible hemorrhagic shock [96], restores cardiac output [97], protects against endothelial glycocalyx shedding with 97% rapid restoration [169], and reduces the incidence of MODS [50, 98]. During resuscitation, we have shown the ALM therapy is neuroprotective at hypotensive pressures (MAPs 47–50 mmHg), which has important implications for sepsis [50,51,52,53, 97, 98]. This data has been use to formulate a Systems Hypothesis of Trauma (SHOT), which may be applicable for sepsis and septic shock [49, 50, 54].

Our proof-of-concept studies in a rat polymicrobial sepsis model [65, 66] and pig lipopolysaccharide (LPS)-endotoxin model [63] have been encouraging. In the pig LPS model, ALM therapy induced a profound and reversible hypotensive state (MAP 47 mmHg) by maintaining CO and lowering SVR with little or no change in tissue O2 delivery [63]. CO was maintained by preserving preload recruitable stroke work and improving VA coupling [63]. Importantly, systemic hypotension is not deleterious if CO, VA coupling and O2 delivery to the tissues are maintained by lowering SVR [51,52,53]. This ALM-induced hypotensive state involves re-setting the CNS-control of O2 delivery to tissue mitochondria, that may be beneficial to sepsis patients [50]. Similarly, ALM therapy induced a reversible hypotensive state in a rat model of polymicrobial sepsis with reduced pulmonary edema and correction of coagulopathy [65]. ALM led to 88% survival after six days, without antibiotics, whereas saline controls died early from inflammatory/immune dysfunction and multiple organ failure [66]. Importantly, the combination of ALM is key to whole body protection whereas the individual actives, A, L or M are not [50]. While appreciating the success rate of translating new drugs to humans is less than 5% [147], further ALM preclinical sepsis studies are required to facilitate translation to human safety trials.

Conclusions

The clinical management of sepsis appears to be at a crossroads and requires a new revolution to better understand how the body responds to an infection, new markers to detect its progression and new system-acting drugs to treat it. The current targeting of any single step along a signalling pathway has not been successful because it ignores the complexity of the system. We argue if the immune-driven, CNS-sympathetic hyperactivation can be suppressed, cardiovascular-endothelial glycocalyx coupling will be improved, O2 delivery will be maintained, and secondary injury, including hyperinflammation and immunosuppression, will be prevented leading to better clinical outcomes. We are developing a systems-based drug in animal sepsis/endotoxemia models, which may confer whole body protection in humans.

Availability of data and materials

The datasets during and/or analysed during the current review can be made available from the corresponding author on reasonable request.

References

  1. Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell. 2006;124:783–801.

    Article  CAS  PubMed  Google Scholar 

  2. Alkaaki A, Al-Radi OO, Khoja A, Alnawawi A, Alnawawi A, Maghrabi A, Altaf A, Aljiffry M. Surgical site infection following abdominal surgery: a prospective cohort study. Can J Surg. 2019;62(2):111–7. https://doi.org/10.1503/cjs.004818. Available from https://www.ncbi.nlm.nih.gov/pubmed/30907567.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Angajala A, Lim S, Phillips JB, Kim JH, Yates C, You Z, Tan M. Diverse roles of mitochondria in immune responses: novel insights into immuno-metabolism. Front Immunol. 2018;9:1605. https://doi.org/10.3389/fimmu.2018.01605. Available from https://www.ncbi.nlm.nih.gov/pubmed/30050539.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Angus DC, van der Poll T. Severe sepsis and septic shock. N Engl J Med. 2013;369:840–51.

    Article  CAS  PubMed  Google Scholar 

  5. Awad S, Allison SP, Lobo DN. The history of 0.9% saline. Clin Nutr. 2008;27:179–88.

    Article  CAS  PubMed  Google Scholar 

  6. Barichello T, Generoso JS, Singer M, Dal-Pizzol F. Biomarkers for sepsis: More than just fever and leukocytosis-a narrative review. Crit Care. 2022;26(1):14. https://doi.org/10.1186/s13054-021-03862-5. Available from https://www.ncbi.nlm.nih.gov/pubmed/34991675.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Barman SM. 2019 ludwig lecture: Rhythms in sympathetic nerve activity are a key to understanding neural control of the cardiovascular system. Am J Physiol Regul Integr Comp Physiol. 2020;318:R191–205.

    Article  CAS  PubMed  Google Scholar 

  8. Baue AE. Multiple, progressive, or sequential systems failure. Arch Surg. 1975;110:779–81.

    Article  CAS  PubMed  Google Scholar 

  9. Baumgarten G, Knuefermann P, Schuhmacher G, Vervölgyi V, von Rappard J, Dreiner U, Fink K, Djoufack C, Hoeft A, Grohé C, Knowlton AA, Meyer R. Toll-like receptor 4, nitric oxide, and myocardial depression in endotoxemia. Shock. 2006;25(1):43–9. https://doi.org/10.1097/01.shk.0000196498.57306.a6.

    Article  CAS  PubMed  Google Scholar 

  10. Bianchi ME. Damps, pamps and alarmins: All we need to know about danger. J Leukoc Biol. 2007;81(1):1–5.

    Article  CAS  PubMed  Google Scholar 

  11. Boehm D, Menke H. A history of fluid management-from “one size fits all” to an individualized fluid therapy in burn resuscitation. Medicina (Kaunas). 2021;57(2):187. https://doi.org/10.3390/medicina57020187. Available from https://www.ncbi.nlm.nih.gov/pubmed/33672128.

    Article  PubMed  Google Scholar 

  12. Bozzay JD, Walker PF, Schechtman DW, Shaikh F, Stewart L, Tribble DR, Bradley MJ, G. Infectious Disease Clinical Research Program Trauma Infectious Disease Outcomes Study. Outcomes of exploratory laparotomy and abdominal infections among combat casualties. J Surg Res. 2021;257:285–93. https://doi.org/10.1016/j.jss.2020.07.075. Available from https://www.ncbi.nlm.nih.gov/pubmed/32866669.

    Article  PubMed  Google Scholar 

  13. Buaboonnam J, Wangkittikal C, Narkbunnam N, Vathana N, Takpradit C, Phuakpet K, Sinlapamongkolkul P, Sanpakit K, Karaketklang K, Pongtanakul B. Outcomes of overt and non-overt disseminated intravascular coagulation using the isth dic scoring system in children: a single-center study. Mediterr J Hematol Infect Dis. 2023;15(1):e2023004. https://doi.org/10.4084/MJHID.2023.004. Available from https://www.ncbi.nlm.nih.gov/pubmed/36660355.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Bui-Thi HD, Gia KT, Electronic address, Le Minh K. Coagulation profiles in patients with sepsis/septic shock identify mixed hypo-hypercoagulation patterns based on rotational thromboelastometry: a prospective observational study. Thromb Res. 2023;227:51–9. https://doi.org/10.1016/j.thromres.2023.05.010. Available from https://www.ncbi.nlm.nih.gov/pubmed/37235948.

    Article  CAS  PubMed  Google Scholar 

  15. Burgelman M, Vandendriessche C, Vandenbroucke RE. Extracellular vesicles: a double-edged sword in sepsis. Pharmaceuticals (Basel). 2021;14(8):829. https://doi.org/10.3390/ph14080829. Available from https://www.ncbi.nlm.nih.gov/pubmed/34451925.

    Article  CAS  PubMed  Google Scholar 

  16. Byrne L, Van Haren F. Fluid resuscitation in human sepsis: time to rewrite history? Ann Intensive Care. 2017;7(1):4. https://doi.org/10.1186/s13613-016-0231-8. Available from https://www.ncbi.nlm.nih.gov/pubmed/28050897.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Cain BS, Harken AH, Meldrum DR. Therapeutic strategies to reduce tnf-alpha mediated cardiac contractile depression following ischemia and reperfusion. J Mol Cell Cardiol. 1999;31(5):931–47.

    Article  CAS  PubMed  Google Scholar 

  18. Cannon WB. Traumatic shock. New York: D. Appleton and Co; 1923.

    Google Scholar 

  19. Caraballoa C, Jaimes F. Organ dysfunction in sepsis: an ominous trajectory from infection to death. Yale J Biol Med. 2019;92:629–40.

    Google Scholar 

  20. Carre JE, Orban JC, Re L, Felsmann K, Iffert W, Bauer M, Suliman HB, Piantadosi CA, Mayhew TM, Breen P, Stotz M, Singer M. Survival in critical illness is associated with early activation of mitochondrial biogenesis. Am J Respir Crit Care Med. 2010;182(6):745–51. https://doi.org/10.1164/rccm.201003-0326OC. Available from https://www.ncbi.nlm.nih.gov/pubmed/20538956.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Cavaillon JM, Singer M, Skirecki T. Sepsis therapies: learning from 30 years of failure of translational research to propose new leads. EMBO Mol Med. 2020;12(4):e10128. https://doi.org/10.15252/emmm.201810128. Available from https://www.ncbi.nlm.nih.gov/pubmed/32176432.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Cecconi M, Evans L, Levy M, Rhodes A. Sepsis and septic shock. Lancet. 2018;392(10141):75–87. https://doi.org/10.1016/S0140-6736(18)30696-2. Available from https://www.ncbi.nlm.nih.gov/pubmed/29937192.

    Article  PubMed  Google Scholar 

  23. Chang JC. Sepsis and septic shock: endothelial molecular pathogenesis associated with vascular microthrombotic disease. Thrombosis J. 2019;17:10.

    Article  Google Scholar 

  24. Cholley B, Le Gall A. Ventriculo-arterial coupling: the comeback? J Thorac Dis. 2016;8(9):2287–9.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Clements TW, Tolonen M, Ball CG, Kirkpatrick AW. Secondary peritonitis and intraabdominal sepsis: an increasingly global disease in search of better systemic therapies. Scand J Surg. 2021;110(2):139–49.

    Article  CAS  PubMed  Google Scholar 

  26. Coccolini F, Sartelli M, Kirkpatrick AW. What do we mean by source control and what are we trying to accomplish with an open abdomen in severe complicated intra-abdominal sepsis? J Trauma Acute Care Surg. 2024;96:e39–40. https://doi.org/10.1097/TA.0000000000004253.

    Article  PubMed  Google Scholar 

  27. Coccolini F, Sartelli M, Sawyer R, Rasa K, Viaggi B, Abu-Zidan F, Soreide K, Hardcastle T, Gupta D, Bendinelli C, Ceresoli M, Shelat VG, Broek RT, Baiocchi GL, Moore EE, Sall I, Podda M, Bonavina L, Kryvoruchko IA, Stahel P, Inaba K, Montravers P, Sakakushev B, Sganga G, Ballestracci P, Malbrain M, Vincent JL, Pikoulis M, Beka SG, Doklestic K, Chiarugi M, Falcone M, Bignami E, Reva V, Demetrashvili Z, Di Saverio S, Tolonen M, Navsaria P, Bala M, Balogh Z, Litvin A, Hecker A, Wani I, Fette A, De Simone B, Ivatury R, Picetti E, Khokha V, Tan E, Ball C, Tascini C, Cui Y, Coimbra R, Kelly M, Martino C, Agnoletti V, Boermeester MA, De’Angelis N, Chirica M, Biffl WL, Ansaloni L, Kluger Y, Catena F, Kirkpatrick AW. Source control in emergency general surgery: Wses, gais, sis-e, sis-a guidelines. World J Emerg Surg. 2023;18(1):41. https://doi.org/10.1186/s13017-023-00509-4. Available from https://www.ncbi.nlm.nih.gov/pubmed/37480129.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Court O, Kumar A, Parrillo JE, Kumar A. Clinical review: myocardial depression in sepsis and septic shock. Crit Care. 2002;6(6):500–8. https://doi.org/10.1186/cc1822. Available from https://www.ncbi.nlm.nih.gov/pubmed/12493071.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Crick F. Of molecules and men. Seattle: University of Washington Press; 1966.

    Google Scholar 

  30. Crile G. Nitrous oxide anaesthesia and a note on anoci-association, a new principle in operative surgery. Surg Gynecol Obstet. 1911;13:170–3.

    Google Scholar 

  31. Curry N, Brohi K. Surgery in traumatic injury and perioperative considerations. Semin Thromb Hemost. 2020;46:73–82.

    Article  PubMed  Google Scholar 

  32. Davenport L, Dobson GP, Letson HL. The role of invasive monitoring in the resuscitation of major burns: a systematic review and meta-analysis. Int J Burns Trauma. 2019;9(2):28–40.

    PubMed  PubMed Central  Google Scholar 

  33. Davenport L, Letson HL, Dobson GP. Immune-inflammatory activation after a single laparotomy in a rat model: effect of adenosine, lidocaine and mg2+ infusion to dampen the stress response. Innate Immun. 2017;23(5):482–94.

    Article  CAS  PubMed  Google Scholar 

  34. Davenport L, Letson HL, Dobson GP. Lung protection after severe thermal burns with adenosine, lidocaine, and magnesium (alm) resuscitation and importance of shams in a rat model. J Care Res. 2023:irad127. https://doi.org/10.1093/jbcr/irad12721.

  35. Davies GR, Lawrence M, Pillai S, Mills GM, Aubrey R, Thomas D, Williams R, Morris K, Evans PA. The effect of sepsis and septic shock on the viscoelastic properties of clot quality and mass using rotational thromboelastometry: a prospective observational study. J Crit Care. 2018;44:7–11. https://doi.org/10.1016/j.jcrc.2017.09.183. Available from https://www.ncbi.nlm.nih.gov/pubmed/28988002.

    Article  PubMed  Google Scholar 

  36. de Montmollin E, Aboab J, Mansart A, Annane D. Bench-to-bedside review: Beta-adrenergic modulation in sepsis. Crit Care. 2009;13(5):230. https://doi.org/10.1186/cc8026. Available from https://www.ncbi.nlm.nih.gov/pubmed/19863760.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Deitch EA. Gut-origin sepsis: evolution of a concept. Surgeon. 2012;10(6):350–6.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Desborough JP. The stress response to trauma and surgery. Br J Anaesth. 2000;85(1):109–17.

    Article  CAS  PubMed  Google Scholar 

  39. Dobson GP. Addressing the global burden of trauma in major surgery. Front Surg. 2015;2:43.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Dobson GP. A chaos of delight: science, religion and myth and the shaping of western thought (first edition 2005), ebook. London: Routledge; 2016.

    Book  Google Scholar 

  41. Dobson GP. Trauma of major surgery: a global problem that is not going away. Int J Surg. 2020;81:47–54.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Dobson GP, Biros E, Letson HL, Morris JL. Living in a hostile world: Inflammation, new drug development and coronavirus. Front Immunol (Inflammation). 2021;11:610131.

    Article  Google Scholar 

  43. Dobson GP, Faggian G, Onorati F, Vinten-Johansen J. Hyperkalemic cardioplegia in adult and pediatric cardiac surgery: end of an era? Front Clin Transl Physiol. 2013;4:1–28.

    Google Scholar 

  44. Dobson GP, Letson HL. Far forward gaps in hemorrhagic shock and prolonged field care: an update of alm fluid therapy for field use. J Spec Oper Med. 2020;20(3):78–84.

    Article  Google Scholar 

  45. Dobson GP, Letson HL, Biros E, Morris JL. Specific pathogen-free (spf) animal status as a variable in biomedical research: have we come full circle? EBioMedicine (Lancet). 2019;41:42–3.

    Article  Google Scholar 

  46. Dobson GP, Letson HL, Sharma R, Sheppard F, Cap AP. Mechanisms of early traumatic-induced coagulopathy (tic): the clot thickens or not? J Trauma Acute Care Surg. 2015;79(2):301–9.

    Article  CAS  PubMed  Google Scholar 

  47. Dobson GP, Morris J, Biros E, Letson HL. Specific pathogen-free animals for civilian and military trauma: a cautionary note in the translation of new drug therapies. Shock. 2020;54(2):232–6.

    Article  CAS  PubMed  Google Scholar 

  48. Dobson GP, Morris JL, Davenport LM, Letson HL. Traumatic-induced coagulopathy as a systems failure: a new window into hemostasis. Semin Thromb Hemost. 2020;46(2):199–214.

    Article  PubMed  Google Scholar 

  49. Dobson GP, Morris JL, Letson HL. Why are bleeding trauma patients still dying? Towards a systems hypothesis of trauma (shot). Front Physiol. 2022;13:99093.

    Article  Google Scholar 

  50. Dobson GP, Morris JL, Letson HL. Adenosine, lidocaine and mg2+ (alm) update: teaching old drugs new tricks. Front Med (Lausanne). 2023;10:1231759.

    Article  PubMed  Google Scholar 

  51. Dobson GP, Morris JL, Letson HL. Alm resuscitation with brain and multi-organ protection for far-forward operations: survival at hypotensive pressures. Mil Med. 2024. In press.

  52. Dobson GP, Morris JL, Letson HL. Pathophysiology of severe burn injuries: new therapeutic opportunities from a systems perspective. J Burn Care Res. 2024. https://doi.org/10.1093/jbcr/irae049. Available from https://www.ncbi.nlm.nih.gov/pubmed/38517382.

  53. Dobson GP, Morris JL, Letson HL. Transforming research to improve therapies for trauma in the twenty-first century: an alternative perspective. Crit Care. 2024;28:135. https://doi.org/10.1186/s13054-024-04913-3.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Dobson GP, Morris JM, Letson HL. Immune dysfunction following severe trauma: a systems failure from the cns to mitochondria. Front Med. 2022;6:968453.

    Article  Google Scholar 

  55. Douglas IS, Alapat PM, Corl KA, Exline MC, Forni LG, Holder AL, Kaufman DA, Khan A, Levy MM, Martin GS, Sahatjian JA, Seeley E, Self WH, Weingarten JA, Williams M, Hansell DM. Fluid response evaluation in sepsis hypotension and shock: a randomized clinical trial. Chest. 2020;158(4):1431–45. https://doi.org/10.1016/j.chest.2020.04.025. Available from https://www.ncbi.nlm.nih.gov/pubmed/32353418.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Dugar S, Choudhary C, Duggal A. Sepsis and septic shock: guideline-based management. Cleve Clin J Med. 2020;87(1):53–64. https://doi.org/10.3949/ccjm.87a.18143. Available from https://www.ncbi.nlm.nih.gov/pubmed/31990655.

    Article  PubMed  Google Scholar 

  57. Evans GH. The abuse of normal salt solution. JAMA. 1911;57:2126–7.

    Article  Google Scholar 

  58. Evans L, Rhodes A, Alhazzani W, Antonelli M, Coopersmith CM, French C, Machado FR, McIntyre L, Ostermann M, Prescott HC, Schorr C, Simpson S, JoostWiersinga W, Alshamsi F, Angus DC, Arabi Y, Azevedo L, Beale R, Beilman G, Belley-Cote E, Burry L, Cecconi M, Centofanti J, Yataco AC, De Waele J, Dellinger RP, Doi K, Du B, Estenssoro E, Ferrer R, Gomersall C, Hodgson C, Moller MH, Iwashyna T, Jacob S, Kleinpell R, Klompas M, Koh Y, Kumar A, Kwizera A, Lobo S, Masur H, McGloughlin S, Mehta S, Mehta Y, Mer M, Nunnally M, Oczkowski S, Osborn T, Papathanassoglou E, Perner A, Puskarich M, Roberts J, Schweickert W, Seckel M, Sevransky J, Sprung CL, Welte T, Zimmerman J, Levy M. Executive summary: surviving sepsis campaign: international guidelines for the management of sepsis and septic shock 2021. Crit Care Med. 2021;49(11):1974–82. https://doi.org/10.1097/CCM.0000000000005357. Available from https://www.ncbi.nlm.nih.gov/pubmed/34643578.

    Article  PubMed  Google Scholar 

  59. Fagan G, Barazanchi A, Coulter G, Leeman M, Hill AG, Eglinton TW. New zealand and australia emergency laparotomy mortality rates compare favourably to international outcomes: a systematic review. ANZ J Surg. 2021;91(12):2583–91. https://doi.org/10.1111/ans.16563. Available from https://www.ncbi.nlm.nih.gov/pubmed/33506977.

    Article  PubMed  Google Scholar 

  60. Funk DJ, Parrillo JE, Kumar A. Sepsis and septic shock: a history. Crit Care Clin. 2009;25(1):83–101, viii. https://doi.org/10.1016/j.ccc.2008.12.003. Available from https://www.ncbi.nlm.nih.gov/pubmed/19268796.

    Article  PubMed  Google Scholar 

  61. Giannoudis PV, Dinopoulos H, Chalidis B, Hall GM. Surgical stress response. Injury. 2006;37(Suppl 5):S3-9.

    Article  PubMed  Google Scholar 

  62. Gofton TE, Young GB. Sepsis-associated encephalopathy. Nat Rev Neurol. 2012;8:557–66.

    Article  CAS  PubMed  Google Scholar 

  63. Granfeldt A, Letson HL, Dobson GP, Shi W, Vinten-Johansen J, Tonnesen E. Adenosine, lidocaine and mg2+ improves cardiac and pulmonary function, induces reversible hypotension and exerts anti-inflammatory effects in an endotoxemic porcine model. Crit Care. 2014;18(6):682. https://doi.org/10.1186/s13054-014-0682-y. Available from https://www.ncbi.nlm.nih.gov/pubmed/25497775.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Greer JR. Pathophysiology of cardiovascular dysfunction in sepsis. BJA Education. 2015;15(6):316–21. https://doi.org/10.1093/bjaceaccp/mkv003.

    Article  Google Scholar 

  65. Griffin MJ, Letson HL, Dobson GP. Adenosine, lidocaine and mg2+ (alm) induces a reversible hypotensive state, reduces lung edema and prevents coagulopathy in the rat model of polymicrobial sepsis. J Trauma Acute Care Surg. 2014;77(3):471–8.

    Article  CAS  PubMed  Google Scholar 

  66. Griffin MJ, Letson HL, Dobson GP. Small-volume adenosine, lidocaine and mg2+ (alm) 4 hour infusion leads to 88% survival after 6 days of experimental sepsis in the rat without antibiotics. Clin Vaccine Immunol. 2016;23(11):863–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Guarracino F, Baldassarri R, Pinsky MR. Ventriculo-arterial decoupling in acutely altered hemodynamic states. Crit Care. 2013;17(2):213–20.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Gyawali K, Ramakrishna K, Dhamoon AS. Sepsis: The evolution in definition, pathophysiology, and management. SAGE Open Med. 2019;7:1–13.

    Article  Google Scholar 

  69. Hailu S, Ayinie A, Amsalu H, Hailu S, Tadesse M, Mamo T, Sagni Y, Mekonen S, Jemal B. Perioperative mortality and its predictors among patients undergoing emergency laparotomy at selected southern ethiopian governmental hospitals, 2022: a multicenter prospective cohort study. Ann Med Surg (Lond). 2023;85(4):746–52. https://doi.org/10.1097/MS9.0000000000000437. Available from https://www.ncbi.nlm.nih.gov/pubmed/37113892.

    Article  PubMed  Google Scholar 

  70. Hajjar RJ, Schwinger RH, Schmidt U, Kim CS, Lebeche D, Doye AA, Gwathmey JK. Myofilament calcium regulation in human myocardium. Circulation. 2000;101(14):1679–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Hall GM. The anesthetic modification of the endocrine and metabolic response to surgery. Ann R Coll Surg Engl. 1985;67:25–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Hasanin A. Fluid responsiveness in acute circulatory failure. J Intensive Care. 2015;3:50.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Hauser CJ, Otterbein LE. Danger signals from mitochondrial damps in trauma and post-injury sepsis. Eur J Trauma Emerg Surg. 2018;44(3):317–24.

    Article  CAS  PubMed  Google Scholar 

  74. Hayakawa M. Pathophysiology of trauma-induced coagulopathy: disseminated intravascular coagulation with the fibrinolytic phenotype. J Intensive Care. 2017;5:14.

    Article  PubMed  PubMed Central  Google Scholar 

  75. Hayakawa M, Asahara T, Henzan N, Murakami H, Yamamoto H, Mukai N, Minami Y, Sugano M, Kubota N, Uegaki S, Kamoshida H, Sawamura A, Nomoto K, Gando S. Dramatic changes of the gut flora immediately after severe and sudden insults. Dig Dis Sci. 2011;56(8):2361–5. https://doi.org/10.1007/s10620-011-1649-3. Available from https://www.ncbi.nlm.nih.gov/pubmed/21384123.

    Article  CAS  PubMed  Google Scholar 

  76. Hemker HC, Al Dieri R, Beguin S. Thrombin generating assays: Accruing clinical evidence. Curr Opinion Hematol. 2004;11:170–5.

    Article  CAS  Google Scholar 

  77. Hollenberg SM, Singer M. Pathophysiology of sepsis-induced cardiomyopathy. Nat Rev Cardiol. 2021;18(6):424–34. https://doi.org/10.1038/s41569-020-00492-2. Available from https://www.ncbi.nlm.nih.gov/pubmed/33473203.

    Article  PubMed  Google Scholar 

  78. Huston JM. The vagus nerve and the inflammatory reflex: Wandering on a new treatment paradigm for systemic inflammation and sepsis. Surg Infect (Larchmt). 2012;13(4):187–93.

    Article  PubMed  Google Scholar 

  79. Hyatt HW, Powers SK. Mitochondrial dysfunction is a common denominator linking skeletal muscle wasting due to disease, aging, and prolonged inactivity. Antioxidants (Basel). 2021;10(4):588.

    Article  CAS  PubMed  Google Scholar 

  80. Ibarz M, Haas LEM, Ceccato A, Artigas A. The critically ill older patient with sepsis: a narrative review. Ann Intensive Care. 2024;14(1):6. https://doi.org/10.1186/s13613-023-01233-7.

    Article  PubMed  PubMed Central  Google Scholar 

  81. Ince C, Mayeux PR, Nguyen T, Gomez H, Kellum JA, Ospina-Tascon GA, Hernandez G, Murray P, De Backer D, A.X. Workgroup. The endothelium in sepsis. Shock. 2016;45(3):259–70. https://doi.org/10.1097/SHK.0000000000000473. Available from https://www.ncbi.nlm.nih.gov/pubmed/26871664.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Janeway CAJ, Medzhitov R. Innate immune recognition. Annu Rev Immunol. 2002;20:197–216.

    Article  CAS  PubMed  Google Scholar 

  83. Joffre J, Hellman J, Ince C, Ait-Oufella H. Endothelial responses in sepsis. Am J Respir Crit Care Med. 2020;202(3):361–70. https://doi.org/10.1164/rccm.201910-1911TR. Available from https://www.ncbi.nlm.nih.gov/pubmed/32101446.

    Article  CAS  PubMed  Google Scholar 

  84. Kanashiro A, Hiroki CH, da Fonseca DM, Birbrair A, Ferreira RG, Bassi GS, Fonseca MD, Kusuda R, Cebinelli GCM, da Silva KP, Wanderley CW, Menezes GB, Alves-Fiho JC, Oliveira AG, Cunha TM, Pupo AS, Ulloa L, Cunha FQ. The role of neutrophils in neuro-immune modulation. Pharmacol Res. 2020;151:104580. https://doi.org/10.1016/j.phrs.2019.104580. Available from https://www.ncbi.nlm.nih.gov/pubmed/31786317.

    Article  CAS  PubMed  Google Scholar 

  85. Kanneganti T-D. Central roles of nlrs and inflammasomes in viral infection. Nat Rev Immunol. 2010;10(10):688–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Kawai T, Akira S. Pathogen recognition with toll-like receptors. Curr Opin Immunol. 2005;17(4):338–44.

    Article  CAS  PubMed  Google Scholar 

  87. Kazune S, Grabovskis A, Cescon C, Strike E, Vanags I. Association between increased arterial stiffness and clinical outcomes in patients with early sepsis: a prospective observational cohort study. Intensive Care Med Exp. 2019;7(1):26. https://doi.org/10.1186/s40635-019-0252-3. Available from https://www.ncbi.nlm.nih.gov/pubmed/31098834.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Keyloun JW, Le TD, Brummel-Ziedins KE, Mclawhorn MM, Bravo MC, Orfeo T, Johnson LS, Moffatt LT, Pusateri AE, Shupp JW, S.S. Group. Inhalation injury is associated with endotheliopathy and abnormal fibrinolytic phenotypes in burn patients: a cohort study. J Burn Care Res. 2022;43(2):432–9. https://doi.org/10.1093/jbcr/irab102. Available from https://www.ncbi.nlm.nih.gov/pubmed/34089618.

    Article  PubMed  Google Scholar 

  89. Khan I, Bai Y, Zha L, Ullah N, Ullah U, Shah SRH, Sun H, Zhang C. Mechanism of the gut microbiota colonization resistance and enteric pathogen infection. Front Cell Infect Microbiol. 2021;11:716299.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Kirkpatrick AW, Coccolini F, Ansaloni L, Roberts DJ, Tolonen M, McKee JL, Leppaniemi A, Faris P, Doig CJ, Catena F, Fabian T, Jenne CN, Chiara O, Kubes P, Manns B, Kluger Y, Fraga GP, Pereira BM, Diaz JJ, Sugrue M, Moore EE, Ren J, Ball CG, Coimbra R, Balogh ZJ, Abu-Zidan FM, Dixon E, Biffl W, MacLean A, Ball I, Drover J, McBeth PB, Posadas-Calleja JG, Parry NG, Di Saverio S, Ordonez CA, Xiao J, Sartelli M, I. Closed Or Open after Laparotomy after Source Control for Severe Complicated Intra-Abdominal Sepsis. Closed or open after source control laparotomy for severe complicated intra-abdominal sepsis (the cool trial): study protocol for a randomized controlled trial. World J Emerg Surg. 2018;13:26. https://doi.org/10.1186/s13017-018-0183-4. Available from https://www.ncbi.nlm.nih.gov/pubmed/29977328.

    Article  PubMed  PubMed Central  Google Scholar 

  91. Kirkpatrick AW, Coccolini F, Tolonen M, Minor S, Catena F, Gois E Jr, Doig CJ, Hill MD, Ansaloni L, Chiarugi M, Tartaglia D, Ioannidis O, Sugrue M, Colak E, Hameed SM, Lampela H, Agnoletti V, McKee JL, Garraway N, Sartelli M, Ball CG, Parry NG, Voght K, Julien L, Kroeker J, Roberts DJ, Faris P, Tiruta C, Moore EE, Ammons LA, Anestiadou E, Bendinelli C, Bouliaris K, Carroll R, Ceresoli M, Favi F, Gurrado A, Rezende-Neto J, Isik A, Cremonini C, Strambi S, Koukoulis G, Testini M, Trpcic S, Pasculli A, Picariello E, Abu-Zidan F, Adeyeye A, Augustin G, Alconchel F, Altinel Y, Hernandez Amin LA, Aranda-Narvaez JM, Baraket O, Biffl WL, Baiocchi GL, Bonavina L, Brisinda G, Cardinali L, Celotti A, Chaouch M, Chiarello M, Costa G, de’Angelis N, De Manzini N, Delibegovic S, Di Saverio S, De Simone B, Dubuisson V, Fransvea P, Garulli G, Giordano A, Gomes C, Hayati F, Huang J, Ibrahim AF, Huei TJ, Jailani RF, Khan M, Luna AP, Malbrain M, Marwah S, McBeth P, Mihailescu A, Morello A, Mulita F, Murzi V, Mohammad AT, Parmar S, Pak A, Wong MP, Pantalone D, Podda M, Puccioni C, Rasa K, Ren J, Roscio F, Gonzalez-Sanchez A, Sganga G, Scheiterle M, Slavchev M, Smirnov D, Tosi L, Trivedi A, Vega JAG, Waledziak M, Xenaki S, Winter D, Wu X, Zakaria AD, Zakaria Z. The unrestricted global effort to complete the cool trial. World J Emerg Surg. 2023;18(1):33. https://doi.org/10.1186/s13017-023-00500-z. Available from https://www.ncbi.nlm.nih.gov/pubmed/37170123.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Koutroulis I, Batabyal R, McNamara B, Ledda M, Hoptay C, Freishtat RJ. Sepsis immunometabolism: from defining sepsis to understanding how energy production affects immune response. Crit Care Explor. 2019;1(11):e0061. https://doi.org/10.1097/CCE.0000000000000061. Available from https://www.ncbi.nlm.nih.gov/pubmed/32166242.

    Article  PubMed  PubMed Central  Google Scholar 

  93. Kumar A, Thota V, Dee L, Olson J, Uretz E, Parrillo JE. Tumor necrosis factor-alpha and interleukin 1-beta are responsible for in vitro myocardial cell depression induced by human septic shock serum. J Exp Med. 1996;83:949–58.

    Article  Google Scholar 

  94. Kumar V. Immunometabolism: another road to sepsis and its therapeutic targeting. Inflammation. 2019;42(3):765–88. https://doi.org/10.1007/s10753-018-0939-8. Available from https://www.ncbi.nlm.nih.gov/pubmed/30506105.

    Article  PubMed  Google Scholar 

  95. Lelubre C, Vincent JL. Mechanisms and treatment of organ failure in sepsis. Nat Rev Nephrol. 2018;14(7):417–27. https://doi.org/10.1038/s41581-018-0005-7. Available from https://www.ncbi.nlm.nih.gov/pubmed/29691495.

    Article  PubMed  Google Scholar 

  96. Letson HL, Biros E, Morris JL, Dobson GP. Alm fluid therapy shifts sympathetic hyperactivity to parasympathetic dominance in the rat model of non-compressible hemorrhagic shock. Shock. 2022;57(2):264–73.

    Article  CAS  PubMed  Google Scholar 

  97. Letson HL, Dobson GP. 3.0% nacl adenosine, lidocaine, mg2+ (alm) bolus and 4 hours ‘drip’ infusion reduces non-compressible hemorrhage by 60% in a rat model. J Trauma Acute Care Surg. 2017;82(6):1063–72.

    Article  CAS  PubMed  Google Scholar 

  98. Letson HL, Granfeldt A, Jensen TH, Mattson TH, Dobson GP. Alm supports a high flow, hypotensive, vasodilatory state with improved o2 delivery and cerebral protection in a pig model of non-compressible hemorrhage. J Surg Res. 2020;253:127–38.

    Article  CAS  PubMed  Google Scholar 

  99. Levi M, Scully M. How i treat disseminated intravascular coagulation. Blood. 2018;131(8):845–54.

    Article  CAS  PubMed  Google Scholar 

  100. Levi M, van der Poll T. Coagulation and sepsis. Thromb Res. 2017;149:38–44. https://doi.org/10.1016/j.thromres.2016.11.007. Available from https://www.ncbi.nlm.nih.gov/pubmed/27886531.

    Article  CAS  PubMed  Google Scholar 

  101. Liang X, FitzGerald GA. Timing of microbes: the circadian rhythm of the gut microbiome. J Biol Rhythm. 2017;32(6):505–15.

    Article  CAS  Google Scholar 

  102. Liu L, Li Y, Chen G, Chen Q. Crosstalk between mitochondrial biogenesis and mitophagy to maintain mitochondrial homeostasis. J Biomed Sci. 2023;30(1):86. https://doi.org/10.1186/s12929-023-00975-7. Available from https://www.ncbi.nlm.nih.gov/pubmed/37821940.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Liu W, Liu T, Zheng Y, Xia Z. Metabolic reprogramming and its regulatory mechanism in sepsis-mediated inflammation. J Inflamm Res. 2023;16:1195–207. https://doi.org/10.2147/JIR.S403778. Available from https://www.ncbi.nlm.nih.gov/pubmed/36968575.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Liu YX, Yu Y, Liu JP, Liu WJ, Cao Y, Yan RM, Yao YM. Neuroimmune regulation in sepsis-associated encephalopathy: the interaction between the brain and peripheral immunity. Front Neurol. 2022;13:892480. https://doi.org/10.3389/fneur.2022.892480. Available from https://www.ncbi.nlm.nih.gov/pubmed/35832175.

    Article  PubMed  PubMed Central  Google Scholar 

  105. Lu Y, Sun Y, Xu K, Shao Y, Saaoud F, Snyder NW, Yang L, Yu J, Wu S, Hu W, Sun J, Wang H, Yang X. Editorial: endothelial cells as innate immune cells. Front Immunol. 2022;13:1035497. https://doi.org/10.3389/fimmu.2022.1035497. Available from https://www.ncbi.nlm.nih.gov/pubmed/36268030.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Luft JH. The structure and properties of the cell surface coat. Int Rev Cytol. 1976;45:291–382.

    Article  CAS  PubMed  Google Scholar 

  107. Luo C, Hu H, Gong J, Zhou Y, Chen Z, Cai S. The value of thromboelastography in the diagnosis of sepsis-induced coagulopathy. Clin Appl Thromb Hemost. 2020;26:1076029620951847. https://doi.org/10.1177/1076029620951847. Available from https://www.ncbi.nlm.nih.gov/pubmed/32870718.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Macdonald S. Fluid resuscitation in patients presenting with sepsis: current insights. Open Access Emerg Med. 2022;14:633–8. https://doi.org/10.2147/OAEM.S363520. Available from https://www.ncbi.nlm.nih.gov/pubmed/36471825.

    Article  PubMed  PubMed Central  Google Scholar 

  109. MacLean LD, Mulligan WG, McLean AP, Duff JH. Patterns of septic shock in man-a detailed study of 56 patients. Ann Surg. 1967;166:543–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Manson J, Cole E, De’Ath HD, Vulliamy P, Meier U, Pennington D, Brohi K. Early changes within the lymphocyte population are associated with the development of multiple organ dysfunction syndrome in trauma patients. Crit Care. 2016;20(1):176. https://doi.org/10.1186/s13054-016-1341-2. Available from https://www.ncbi.nlm.nih.gov/pubmed/27268230.

    Article  PubMed  PubMed Central  Google Scholar 

  111. Marik PE, Lemson J. Fluid responsiveness: an evolution of our understanding. Br J Anaesth. 2014;112(4):617–20.

    Article  CAS  PubMed  Google Scholar 

  112. Matzinger P. The evolution of the danger theory. Expert Rev Clin Immunol. 2012;8(4):311–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Mayer EA, Tillisch K, Gupta A. Gut/brain axis and the microbiota. J Clin Invest. 2015;125(3):926–38.

    Article  PubMed  PubMed Central  Google Scholar 

  114. McBride MA, Owen AM, Stothers CL, Hernandez A, Luan L, Burelbach KR, Patil TK, Bohannon JK, Sherwood ER, Patil NK. The metabolic basis of immune dysfunction following sepsis and trauma. Front Immunol. 2020;11:1043. https://doi.org/10.3389/fimmu.2020.01043. Available from https://www.ncbi.nlm.nih.gov/pubmed/32547553.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Meng X, Ao L, Song Y, Raeburn CD, Fullerton DA, Harken AH. Signaling for myocardial depression in hemorrhagic shock: Roles of toll-like receptor 4 and p55 tnf-alpha receptor. Am J Physiol Regul Integr Comp Physiol. 2005;288(3):R600-606. https://doi.org/10.1152/ajpregu.00182.2004. Available from https://www.ncbi.nlm.nih.gov/pubmed/15514106.

    Article  CAS  PubMed  Google Scholar 

  116. Metchnikoff E. Lectures on the comparative pathology of inflammation, delivered at the pasteur institute in 1891. New York: Dover Publications (1968); 1891.

    Google Scholar 

  117. Morris JL, Letson HL, Gillman R, Hazratwala K, Wilkinson M, McEwen P, Dobson GP. The cns theory of osteoarthritis: opportunities beyond the joint. Semin Arthritis Rheum. 2019;49:331–6.

    Article  CAS  PubMed  Google Scholar 

  118. Morris JL, Letson HL, McEwen P, Biros E, Dlaska C, Hazratwala K, Wilkinson M, Dobson GP. Intra-articular adenosine, lidocaine and magnesium (alm) solution decreases post-operative fibrosis in a knee implant model. Transl Med Commun. 2021;6(4):1–13.

    Google Scholar 

  119. Morris JL, McEwen PC, Letson HL, Dobson GP. Adenosine, lidocaine and magnesium (alm) therapy modulates early sex-specific inflammatory and immune responses following experimental anterior cruciate ligament rupture and reconstruction. Transl Med Commun. 2023;8(1):14. https://doi.org/10.1186/s41231-023-00148-6.

    Article  Google Scholar 

  120. Murphy JB. Iv. Treatment of perforative peritonitis. Ann Surg. 1908;47(6):870–2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Myburgh J. Patient-centered outcomes and resuscitation fluids. N Engl J Med. 2018;378(9):862–3. https://doi.org/10.1056/NEJMe1800449. Available from https://www.ncbi.nlm.nih.gov/pubmed/29485927.

    Article  PubMed  Google Scholar 

  122. Nasir N, Jamil B, Siddiqui S, Talat N, Khan FA, Hussain R. Mortality in sepsis and its relationship with gender. Pak J Med Sci. 2015;31(5):1201–6. https://doi.org/10.12669/pjms.315.6925. Available from https://www.ncbi.nlm.nih.gov/pubmed/26649014.

    Article  PubMed  PubMed Central  Google Scholar 

  123. Neubauer K, Zieger B. Endothelial cells and coagulation. Cell Tissue Res. 2022;387(3):391–8. https://doi.org/10.1007/s00441-021-03471-2. Available from https://www.ncbi.nlm.nih.gov/pubmed/34014399.

    Article  CAS  PubMed  Google Scholar 

  124. Nwafor DC, Brichacek AL, Mohammad AS, Griffith J, Luke-Wold BP, Benkovic S, Geldenhuys WJ, Lockman PR, Brown CM. Targeting the blood-brain barrier to prevent sepsis-associated cognitive impairment. J Centr Nerv Syst Dis. 2019;11:1–14.

    Google Scholar 

  125. Okamoto K, Tamura T, Sawatsubashi Y. Sepsis and disseminated intravascular coagulation. J Intensive Care. 2016;4:23. https://doi.org/10.1186/s40560-016-0149-0. Available from https://www.ncbi.nlm.nih.gov/pubmed/27011792.

    Article  PubMed  PubMed Central  Google Scholar 

  126. Okamoto T, Tanigami H, Suzuki K, Shimaoka M. Thrombomodulin: a bifunctional modulator of inflammation and coagulation in sepsis. Crit Care Res Pract. 2012;2012:614545. https://doi.org/10.1155/2012/614545. Available from https://www.ncbi.nlm.nih.gov/pubmed/22482044.

    Article  PubMed  PubMed Central  Google Scholar 

  127. Opal SM. The evolution of the understanding of sepsis, infection, and the host response: a brief history. Crit Care Clin. 2009;25(4):637–63, vii. https://doi.org/10.1016/j.ccc.2009.08.007. Available from https://www.ncbi.nlm.nih.gov/pubmed/19892245.

    Article  PubMed  Google Scholar 

  128. Osler W. The evolution of modern medicine. New Haven: Yale University Press; 1921.

    Google Scholar 

  129. Panigrahy D, Gilligan MM, Huang S, Gartung A, Cortes-Puch I, Sime PJ, Phipps RP, Serhan CN, Hammock BD. Inflammation resolution: a dual-pronged approach to averting cytokine storms in covid-19? Cancer Metastasis Rev. 2020;39(2):337–40. https://doi.org/10.1007/s10555-020-09889-4. Available from https://www.ncbi.nlm.nih.gov/pubmed/32385712.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Parker MM, Shelhamer JH, Bacharach SL, Green MV, Natanson C, Frederick TM, Damske BA, Parrillo JE. Profound but reversible myocardial depression in patients with septic shock. Ann Intern Med. 1984;100(4):483–90.

    Article  CAS  PubMed  Google Scholar 

  131. Parrillo JE, Burch C, Shelhamer JH, Parker MM, Natanson C, Schuette W. A circulating myocardial depressant substance in humans with septic shock. Septic shock patients with a reduced ejection fraction have a circulating factor that depresses in vitro myocardial cell performance. J Clin Invest. 1985;76(4):1539–53. https://doi.org/10.1172/JCI112135. Available from https://www.ncbi.nlm.nih.gov/pubmed/4056039.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Pavlov VA, Tracey KJ. Neural regulators of innate immune responses and inflammation. Cell Mol Life Sci. 2004;61:2322–31.

    Article  CAS  PubMed  Google Scholar 

  133. Picard M, McManus MJ, Gray JD, Nasca C, Moffat C, Kopinski PK, Seifert EL, McEwen BS, Wallace DC. Mitochondrial functions modulate neuroendocrine, metabolic, inflammatory, and transcriptional responses to acute psychological stress. Proc Natl Acad Sci U S A. 2015;112(48):E6614-6623. https://doi.org/10.1073/pnas.1515733112. Available from https://www.ncbi.nlm.nih.gov/pubmed/26627253.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Piccinini AM, Midwood KS. Dampening inflammation by modulating tlr signalling. Mediators Inflamm. 2010;2010:1–21.

    Article  Google Scholar 

  135. Pinsky MR, Guarracino F. How to assess ventriculoarterial coupling in sepsis. Curr Opin Crit Care. 2020;26(3):313–8. https://doi.org/10.1097/MCC.0000000000000721. Available from https://www.ncbi.nlm.nih.gov/pubmed/32348096.

    Article  PubMed  PubMed Central  Google Scholar 

  136. Pinto MY, Frois AO, Weber D. A retrospective cohort study on one-year mortality following emergency laparotomy: a tertiary centre experience from western australia. Cureus. 2023;15(12):e50718. https://doi.org/10.7759/cureus.50718. Available from https://www.ncbi.nlm.nih.gov/pubmed/38234926.

    Article  PubMed  PubMed Central  Google Scholar 

  137. Rabuel C, Mebazaa A. Septic shock: a heart story since the 1960s. Intensive Care Med. 2006;32:799–807.

    Article  CAS  PubMed  Google Scholar 

  138. Razazi K, Boissier F, Surenaud M, Bedet A, Seemann A, Carteaux G, de Prost N, Brun-Buisson C, Hue S, MekontsoDessap A. A multiplex analysis of sepsis mediators during human septic shock: a preliminary study on myocardial depression and organ failures. Ann Intensive Care. 2019;9(1):64. https://doi.org/10.1186/s13613-019-0538-3. Available from https://www.ncbi.nlm.nih.gov/pubmed/31165286.

    Article  PubMed  PubMed Central  Google Scholar 

  139. Riva G, Nasillo V, Luppi M, Tagliafico E, Trenti T. Linking covid-19, monocyte activation and sepsis: Mdw, a novel biomarker from cytometry. EBioMedicine. 2022;75:103754. https://doi.org/10.1016/j.ebiom.2021.103754. Available from https://www.ncbi.nlm.nih.gov/pubmed/34922322.

    Article  CAS  PubMed  Google Scholar 

  140. Rivers E, Nguyen B, Havstad S, Ressler J, Muzzin A, Knoblich B, Peterson E, Tomlanovich M, F.t.e.g.-d.t.c. group. Early goal-directed therapy in the treatment of severe sepsis and septic shock. N Engl J Med. 2001;345(19):1368–77.

    Article  CAS  PubMed  Google Scholar 

  141. Roh JS, Sohn DH. Damage-associated molecular patterns in inflammatory diseases. Immune Netw. 2018;18(4):e27.

    Article  PubMed  PubMed Central  Google Scholar 

  142. Rudd KE, Johnson SC, Agesa KM, Shackelford KA, Tsoi D, Kievlan DR, Colombara DV, Ikuta KS, Kissoon N, Finfer S, Fleischmann-Struzek C, Machado FR, Reinhart KK, Rowan K, Seymour CW, Watson RS, West TE, Marinho F, Hay SI, Lozano R, Lopez AD, Angus DC, Murray CJL, Naghavi M. Global, regional, and national sepsis incidence and mortality, 1990–2017: analysis for the global burden of disease study. Lancet. 2020;395(10219):200–11. https://doi.org/10.1016/S0140-6736(19)32989-7. Available from https://www.ncbi.nlm.nih.gov/pubmed/31954465.

    Article  PubMed  PubMed Central  Google Scholar 

  143. Santacroce E, D’Angerio M, Ciobanu AL, Masini L, Lo Tartaro D, Coloretti I, Busani S, Rubio I, Meschiari M, Franceschini E, Mussini C, Girardis M, Gibellini L, Cossarizza A, De Biasi S. Advances and challenges in sepsis management: modern tools and future directions. Cells. 2024;13(5):439. https://doi.org/10.3390/cells13050439. Available from https://www.ncbi.nlm.nih.gov/pubmed/38474403.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Schröder J, Kahlke V, Staubach K-H, Zabel P, Stüber F. Gender differences in human sepsis. Arch Surg. 1998;133(11):1200–5.

    Article  PubMed  Google Scholar 

  145. Schulz R, Panas DL, Catena R, Moncada S, Olley PM, Lopaschuk GD. The role of nitric oxide in cardiac depression induced by interleukin-1beta and tumour necrosis factor-alpha. Br J Pharmacol. 1995;114:27–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Sekino N, Selim M, Shehadah A. Sepsis-associated brain injury: Underlying mechanisms and potential therapeutic strategies for acute and long-term cognitive impairments. J Neuroinflammation. 2022;19(1):101. https://doi.org/10.1186/s12974-022-02464-4. Available from https://www.ncbi.nlm.nih.gov/pubmed/35488237.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Seyhan AA. Lost in translation: the valley of death across preclinical and clinical divide – identification of problems and overcoming obstacles. Transl Med Commun. 2019;4(1):1–9. https://doi.org/10.1186/s41231-019-0050-7.

    Article  Google Scholar 

  148. Seymour CW, Kennedy JN, Wang S, Chang CH, Elliott CF, Xu Z, Berry S, Clermont G, Cooper G, Gomez H, Huang DT, Kellum JA, Mi Q, Opal SM, Talisa V, van der Poll T, Visweswaran S, Vodovotz Y, Weiss JC, Yealy DM, Yende S, Angus DC. Derivation, validation, and potential treatment implications of novel clinical phenotypes for sepsis. JAMA. 2019;321(20):2003–17. https://doi.org/10.1001/jama.2019.5791. Available from https://www.ncbi.nlm.nih.gov/pubmed/31104070.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Shapiro L, Scherger S, Franco-Paredes C, Gharamti A, Henao-Martinez AF. Anakinra authorized to treat severe coronavirus disease 2019; sepsis breakthrough or time to reflect? Front Microbiol. 2023;14:1250483. https://doi.org/10.3389/fmicb.2023.1250483. Available from https://www.ncbi.nlm.nih.gov/pubmed/37928695.

    Article  PubMed  PubMed Central  Google Scholar 

  150. Shapiro NI, Douglas IS, Brower RG, Brown SM, Exline MC, Ginde AA, Gong MN, Grissom CK, Hayden D, Hough CL, Huang W, Iwashyna TJ, Jones AE, Khan A, Lai P, Liu KD, Miller CD, Oldmixon K, Park PK, Rice TW, Ringwood N, Semler MW, Steingrub JS, Talmor D, Thompson BT, Yealy DM, Self WH. Early restrictive or liberal fluid management for sepsis-induced hypotension. National heart, lung blood institute, prevention early treatment of acute lung injury clinical trials, network. N Engl J Med. 2023;388(6):499–510. https://doi.org/10.1056/NEJMoa2212663. Available from https://www.ncbi.nlm.nih.gov/pubmed/36688507.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Sharawy N. Vasoplegia in septic shock: do we really fight the right enemy? J Crit Care. 2014;29:83–7.

    Article  CAS  PubMed  Google Scholar 

  152. Silva EE, Skon-Hegg C, Badovinac VP, Griffith TS. The calm after the storm: Implications of sepsis immunoparalysis on host immunity. J Immunol. 2023;211(5):711–9. https://doi.org/10.4049/jimmunol.2300171. Available from https://www.ncbi.nlm.nih.gov/pubmed/37603859.

    Article  CAS  PubMed  Google Scholar 

  153. Silverman MN, Pearce BD, Biron CA, Miller AH. Immune modulation of the hypothalamic-pituitary-adrenal (hpa) axis during viral infection. Viral Immunol. 2005;18(1):41–78.

    Article  CAS  PubMed  Google Scholar 

  154. Sincavage J, Msosa VJ, Katete C, Purcell LN, Charles A. Postoperative complications and risk of mortality after laparotomy in a resource-limited setting. J Surg Res. 2021;260:428–35. https://doi.org/10.1016/j.jss.2020.11.017. Available from https://www.ncbi.nlm.nih.gov/pubmed/33272596.

    Article  PubMed  Google Scholar 

  155. Skei NV, Nilsen TIL, Mohus RM, Prescott HC, Lydersen S, Solligard E, Damas JK, Gustad LT. Trends in mortality after a sepsis hospitalization: a nationwide prospective registry study from 2008 to 2021. Infection. 2023;51(6):1773–86. https://doi.org/10.1007/s15010-023-02082-z. Available from https://www.ncbi.nlm.nih.gov/pubmed/37572240.

    Article  PubMed  PubMed Central  Google Scholar 

  156. Solari E, Marcozzi C, Negrini D, Moriondo A. Interplay between gut lymphatic vessels and microbiota. Cells. 2021;10(10):2584. https://doi.org/10.3390/cells10102584. Available from https://www.ncbi.nlm.nih.gov/pubmed/34685564.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Spragge F, Bakkeren E, Jahn MT, Araujo BN, E, Pearson CF, Wang X, Pankhurst L, Cunrath O, Foster KR,. Microbiome diversity protects against pathogens by nutrient blocking. Science. 2023;382(6676):eadj3502. https://doi.org/10.1126/science.adj3502. Available from https://www.ncbi.nlm.nih.gov/pubmed/38096285.

    Article  CAS  PubMed  Google Scholar 

  158. Steeb G, Wang Y-Z, Siegrist B, O’Leary JP. Infections within the peritoneal cavity: a historical perspective. Am Surg. 2000;66(2):98–104.

    Article  CAS  PubMed  Google Scholar 

  159. Suzuki T, Suzuki Y, Okuda J, Kurazumi T, Suhara T, Ueda T, Nagata H, Morisaki H. Sepsis-induced cardiac dysfunction and beta-adrenergic blockade therapy for sepsis. J Intensive Care. 2017;5:22. https://doi.org/10.1186/s40560-017-0215-2. Available from https://www.ncbi.nlm.nih.gov/pubmed/28270914.

    Article  PubMed  PubMed Central  Google Scholar 

  160. Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell. 2020;140(6):805–20.

    Article  Google Scholar 

  161. Tavernier B, Garrigue D, Boulle C, Vallet B, Adnet P. Myofilament calcium sensitivity is decreased in skinned cardiac fibres of endotoxin-treated rabbits. Cardiovasc Res. 1998;38:472–9.

    Article  CAS  PubMed  Google Scholar 

  162. Tengberg LT, Cihoric M, Foss NB, Bay-Nielsen M, Gogenur I, Henriksen R, Jensen TK, Tolstrup MB, Nielsen LB. Complications after emergency laparotomy beyond the immediate postoperative period - a retrospective, observational cohort study of 1139 patients. Anaesthesia. 2017;72(3):309–16. https://doi.org/10.1111/anae.13721. Available from https://www.ncbi.nlm.nih.gov/pubmed/27809332.

    Article  CAS  PubMed  Google Scholar 

  163. Thachil J. The elusive diagnosis of disseminated intravascular coagulation: does a diagnosis of dic exist anymore? Semin Thromb Hemost. 2019;45:100–7.

    Article  PubMed  Google Scholar 

  164. Thurairajah K, Briggs GD, Balogh ZJ. The source of cell-free mitochondrial DNA in trauma and potential therapeutic strategies. Eur J Trauma Emerg Surg. 2018;44(3):325–34. https://doi.org/10.1007/s00068-018-0954-3. Available from https://www.ncbi.nlm.nih.gov/pubmed/29633007.

    Article  PubMed  PubMed Central  Google Scholar 

  165. Tian C, Wang K, Zhao M, Cong S, Di X, Li R. Extracellular vesicles participate in the pathogenesis of sepsis. Front Cell Infect Microbiol. 2022;12:1018692. https://doi.org/10.3389/fcimb.2022.1018692. Available from https://www.ncbi.nlm.nih.gov/pubmed/36579343.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Tillisch K. The effects of gut microbiota on cns function in humans. Gut Microbes. 2014;5(3):404–10.

    Article  PubMed  PubMed Central  Google Scholar 

  167. Toh CH, Hoots WK, S.S.C.o.D.I.C.o.t. ISTH. The scoring system of the scientific and standardisation committee on disseminated intravascular coagulation of the international society on thrombosis and haemostasis: a 5-year overview. J Thromb Haemost. 2007;5(3):604–6. https://doi.org/10.1111/j.1538-7836.2007.02313.x. Available from https://www.ncbi.nlm.nih.gov/pubmed/17096704.

    Article  CAS  PubMed  Google Scholar 

  168. Torp MK, Stenslokken KO, Vaage J. When our best friend becomes our worst enemy: The mitochondrion in trauma, surgery, and critical illness. J Intensive Care Med. 2024:8850666241237715. https://doi.org/10.1177/08850666241237715. Available from https://www.ncbi.nlm.nih.gov/pubmed/38505947.

  169. Torres Filho IP, Torres LN, Salgado C, Dubick MA. Novel adjunct drugs reverse endothelial glycocalyx damage after hemorrhagic shock in rats. Shock. 2017;48(5):583–9.

    Article  CAS  PubMed  Google Scholar 

  170. Tracey KJ. The inflammatory reflex. Nature. 2020;420:853–9.

    Article  Google Scholar 

  171. Uchimido R, Schmidt EP, Shapiro NI. The glycocalyx: a novel diagnostic and therapeutic target in sepsis. Crit Care. 2019;23(1):16.

    Article  PubMed  PubMed Central  Google Scholar 

  172. Unar A, Bertolino L, Patauner F, Gallo R, Durante-Mangoni E. Pathophysiology of disseminated intravascular coagulation in sepsis: a clinically focused overview. Cells. 2023;12(17):2120. https://doi.org/10.3390/cells12172120. Available from https://www.ncbi.nlm.nih.gov/pubmed/37681852.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Vijay K. Toll-like receptors in immunity and infl ammatory diseases: past, present, and future. Int Immunopharmacol. 2018;59:391–412.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Weber B, Henrich D, Hildebrand F, Marzi I, Leppik L. The roles of extracellular vesicles in sepsis and systemic inflammatory response syndrome. Shock. 2023;59(2):161–72. https://doi.org/10.1097/SHK.0000000000002010. Available from https://www.ncbi.nlm.nih.gov/pubmed/36730865.

    Article  CAS  PubMed  Google Scholar 

  175. Weisel RD. Myocardial stunning after coronary bypass surgery. J Card Surg. 1993;8:242–4.

    Article  CAS  PubMed  Google Scholar 

  176. Welling H, Henriksen HH, Gonzalez-Rodriguez ER, Stensballe J, Huzar TF, Johansson PI, Wade CE. Endothelial glycocalyx shedding in patients with burns. Burns. 2020;46(2):386–93. https://doi.org/10.1016/j.burns.2019.05.009. Available from https://www.ncbi.nlm.nih.gov/pubmed/31866179.

    Article  PubMed  Google Scholar 

  177. Wiersinga WJ, van der Poll T. Immunopathophysiology of human sepsis. EBioMedicine. 2022;86:104363. https://doi.org/10.1016/j.ebiom.2022.104363. Available from https://www.ncbi.nlm.nih.gov/pubmed/36470832.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Wilhelm G, Mertowska P, Mertowski S, Przysucha A, Struzyna J, Grywalska E, Torres K. The crossroads of the coagulation system and the immune system: Interactions and connections. Int J Mol Sci. 2023;24(16):12563. https://doi.org/10.3390/ijms241612563. Available from https://www.ncbi.nlm.nih.gov/pubmed/37628744.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Williams B, Zou L, Pittet JF, Chao W. Sepsis-induced coagulopathy: a comprehensive narrative review of pathophysiology, clinical presentation, diagnosis, and management strategies. Anesth Analg. 2024. https://doi.org/10.1213/ANE.0000000000006888. Available from https://www.ncbi.nlm.nih.gov/pubmed/38324297.

  180. Wilson RF, Thal AP, Kindling PH, Grifka T, Ackerman E. Hemodynamic measurements in septic shock. Ann Surg. 1965;91:121–9.

    Google Scholar 

  181. Wu C, Lu W, Zhang Y, Zhang G, Shi X, Hisada Y, Grover SP, Zhang X, Li L, Xiang B, Shi J, Li XA, Daugherty A, Smyth SS, Kirchhofer D, Shiroishi T, Shao F, Mackman N, Wei Y, Li Z. Inflammasome activation triggers blood clotting and host death through pyroptosis. Immunity. 2019;50(6):1401-1411 e1404. https://doi.org/10.1016/j.immuni.2019.04.003. Available from https://www.ncbi.nlm.nih.gov/pubmed/31076358.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Xu C, Yi C, Wang H, Bruce IC, Xia Q. Mitochondrial nitric oxide synthase participates in septic shock myocardial depression by nitric oxide overproduction and mitochondrial permeability transition pore opening. Shock. 2012;37(1):110–5. https://doi.org/10.1097/SHK.0b013e3182391831. Available from https://www.ncbi.nlm.nih.gov/pubmed/21993446.

    Article  CAS  PubMed  Google Scholar 

  183. Young GB. Commentary on posterior reversible encephalopathy syndrome and sepsis-associated encephalopathy. Neurocrit Care. 2022;37(1):8–9. https://doi.org/10.1007/s12028-022-01443-0. Available from https://www.ncbi.nlm.nih.gov/pubmed/35133606.

    Article  PubMed  Google Scholar 

  184. Young JD. The heart and circulation in severe sepsis. Br J Anaesth. 2004;93(1):114–20. https://doi.org/10.1093/bja/aeh171. Available from https://www.ncbi.nlm.nih.gov/pubmed/15121730.

    Article  CAS  PubMed  Google Scholar 

  185. Zeng Y, Tarbell JM. The adaptive remodeling of endothelial glycocalyx in response to fluid shear stress. PLoS ONE. 2014;9(1):e86249.

    Article  PubMed  PubMed Central  Google Scholar 

  186. Zizzo G, Cohen PL. Il-17 stimulates differentiation of human anti-inflammatory macrophages and phagocytosis of apoptotic neutrophils in response to il-10 and glucocorticoids. J Immunol. 2013;190(10):5237–46.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors would like to thank the US Department of Defense and James Cook University’s College of Medicine and Dentistry for their continued support.

Funding

No funding.

Author information

Authors and Affiliations

Authors

Contributions

Concept (GPD), data collection (GPD, JLM, HLL), data analyses and interpretation (GPD, JLM, HLL), manuscript preparation (GPD, JLM, HLL) and editing (GPD, JLM, HLL). hayley.letson@jcu.edu.au, Jodie.morris1@jcu.edu.au.

Corresponding author

Correspondence to Geoffrey P. Dobson.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

GPD is the sole inventor of the ALM concept for cardiac surgery, trauma and sepsis. JLM and HLL have no conflicts to declare.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dobson, G.P., Letson, H.L. & Morris, J.L. Revolution in sepsis: a symptoms-based to a systems-based approach?. J Biomed Sci 31, 57 (2024). https://doi.org/10.1186/s12929-024-01043-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12929-024-01043-4

Keywords