Next Article in Journal
Feeding Cactus (Opuntia stricta [Haw.] Haw.) Cladodes as a Partial Substitute for Elephant Grass (Pennisetum purpureum Schum.) Induces Beneficial Changes in Milk Fatty Acid Composition of Dairy Goats Fed Full-Fat Corn Germ
Previous Article in Journal
Acid Whey from Industrial Greek Strained Yoghurt: Effect of the Kind of Milk and the Way of Straining on Its Composition and Processing by Nanofiltration
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Calci-Inflammatory Network: A Paradigm Shift in Understanding Milk Fever

Department of Agriculture, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, AB T6G 2P5, Canada
Dairy 2025, 6(3), 22; https://doi.org/10.3390/dairy6030022
Submission received: 11 March 2025 / Revised: 11 April 2025 / Accepted: 25 April 2025 / Published: 28 April 2025
(This article belongs to the Section Dairy Animal Health)

Abstract

:
This review highlights a paradigm shift in our understanding of hypocalcemia during milk fever by introducing the concept of the Calci-Inflammatory Network. Traditionally viewed as a pathological deficiency necessitating rapid correction (e.g., through calcium borogluconate infusions or dietary adjustments like dietary cation-anion difference), periparturient hypocalcemia is reinterpreted here as an adaptive, protective response. Within this new framework, reduced circulating calcium levels may help temper systemic inflammation by limiting lipopolysaccharide (LPS) aggregation and curbing excessive macrophage activation. The review discusses how calcium signaling, the calcium-sensing receptor (CaSR), and immune cell functions adapt under hypocalcemic conditions to modulate inflammatory processes. This integrated perspective not only redefines the role of hypocalcemia but also proposes the Calci-Inflammatory Network as a novel concept through which we can understand how changes in calcium homeostasis mitigate inflammatory cascades—potentially lowering the incidence of periparturient diseases and enhance overall cow health and farm productivity. Future research should investigate the long-term effects of hypocalcemia, the environmental influences on this Calci-Inflammatory Network, and their collective impact on disease susceptibility and inflammation.

1. Introduction

Milk fever, or periparturient hypocalcemia, has long been recognized as a critical metabolic disorder affecting dairy cows during the transition period. Traditionally, this condition has been understood as a direct consequence of a significant drop in blood calcium levels [1,2], leading to the widespread adoption of preventive and therapeutic strategies focused on managing dietary calcium prior to parturition and dietary adjustments. Approaches such as dietary cation-anion difference (DCAD) manipulation [3], calcium boluses, and subcutaneous or intravenous calcium borogluconate have been developed to counteract hypocalcemia, reflecting a reductionist view that equates milk fever primarily with calcium deficiency [4].
However, despite the widespread use of these interventions, milk fever remains a persistent challenge in dairy herds across North America [2,5] and globally [6,7], with varying proportions of cows—particularly based on parity—still affected by the disorder. The continued presence of milk fever in nearly all dairy herds, despite the apparent success of traditional strategies, calls for a critical reassessment of the foundational assumptions underlying current prevention and management approaches.
If correcting calcium deficiency were indeed the central solution, the persistence of the disorder suggests that these strategies may fail to address the full complexity of the condition [2,8].
This paradox highlights a potential gap in our understanding of milk fever, indicating that the condition might not be as straightforward as previously thought. Emerging research, notably the hypothesis proposed by Ametaj and his team [9,10,11], challenges the conventional paradigm by suggesting that hypocalcemia during milk fever may not merely represent a pathological deficiency but rather an evolutionary adaptive physiological response aimed at mitigating systemic inflammation. This shift in perspective encourages us to reconsider hypocalcemia not as a problem to be corrected but as a potential protective mechanism within the broader context of the cow’s immune response.
Building on this adaptive hypothesis, this review introduces a novel concept—“The Calci-Inflammatory Network”—which proposes that fluctuations in circulating calcium levels, sensed in part by the calcium-sensing receptor (CaSR) on immune cells, play a pivotal role in orchestrating inflammatory processes. Rather than viewing transient hypocalcemia purely as a deficiency, the Calci-Inflammatory Network proposes that controlled reductions in calcium concentration can help modulate the intensity of immune activation and cytokine release, potentially averting excessive inflammation. By framing hypocalcemia as a physiological buffer rather than a pathological anomaly, this perspective offers a fresh lens through which to understand how calcium dynamics can influence both metabolic and immunological outcomes during the periparturient period.
In this review, I will revisit the historical context of milk fever management, critically analyzing why conventional approaches have often fallen short of fully resolving the condition. I will then discuss the growing body of evidence supporting the concept that hypocalcemia may play an adaptive, anti-inflammatory role, particularly in the context of endotoxemia and inflammation. By examining the physiological and molecular mechanisms underlying calcium homeostasis and immune regulation, I aim to deepen the understanding of milk fever. Our objective is not to introduce new management strategies but to refine and expand the current knowledge of milk fever from an integrative perspective.

2. Evolving Hypotheses of Milk Fever: A Historical and Contemporary Overview

Milk fever, also known as parturient paresis, has long been recognized as one of the most complex and puzzling diseases in veterinary medicine, particularly affecting dairy cows during the postparturient period. Since its identification in the 18th century, the condition has prompted numerous hypotheses and research endeavors aimed at unraveling its etiology, reflecting ongoing challenges in veterinary science. The evolution of scientific thought surrounding milk fever is well documented in the seminal review by Hibbs in 1950 [12], which provides a comprehensive historical perspective on the different hypotheses proposed until that time. Despite extensive research efforts, many of these early postulates were eventually proven to be incorrect or incomplete, and new hypotheses continue to emerge as our understanding of the disease deepens.

2.1. Historical Context and Early Theories

2.1.1. Hibbs Review and Subsequent Hypotheses

One of the most comprehensive reviews on milk fever was authored by Hibbs [12]. By that time, milk fever had become a significant focus of research, and by the 1950s, around 30 different hypotheses had been proposed to explain the condition. The disease was mentioned for the first time in 1793 by Eberhardt [13]. The proposed hypotheses ranged widely—encompassing general inflammation, nervous system disorders, circulatory issues, cerebral anemia and congestion, apoplexy, thrombosis, fat embolism, spinal trauma, general infections, bacterial infections of uterine or mammary origin, anaphylaxis, mammary-neurasthenia, dehydration, auto-intoxication, impaired tissue oxidation, excess oxytocin post-parturition, faulty protein metabolism, ovarian dysfunction, hyperactivity of the anterior pituitary, disturbed cholesterol metabolism, hyperadrenalinemia, magnesium narcosis, alkalosis, acidosis, auto-asphyxiation, and hypoglycemia [12]. Although diverse, many of these theories have not stood the test of time.

2.1.2. Development of the Hypocalcemia Theory

The first authors to propose a parathyroid deficiency and hypocalcemia in dairy cows affected by milk fever were Dryerre and Greig in 1925 [14]. Based on clinical observations and speculative reasoning, they hypothesized that udder insufflation would stimulate the secretion of adrenalin to “oxidize toxins”. However, the nature of these toxins was not elaborated, and the hypothesis was soon discarded when experiments by Auger [15] failed to detect increased blood pressure indicative of elevated adrenalin. A pivotal development came with the work of Little and Write (1925) [16], who demonstrated a significant decrease in blood calcium concentrations in cows with clinical milk fever, thereby establishing hypocalcemia as a key feature of the disease. Almost a decade later, Dryerre and Greig (1935) [17] demonstrated that subcutaneous or intramuscular injections of calcium boro-gluconate improved clinical signs, reinforcing that an acute drop in blood calcium—precipitated by the sudden onset of lactation—was central to the pathophysiology. Although these findings lowered milk fever incidence and led to widely adopted therapeutic measures, the hypocalcemia hypothesis alone could not explain every aspect of the disease, driving further research into endocrine and metabolic imbalances that underlie calcium regulation.

2.1.3. The DCAD and Potassium Hypothesis: A Shift in Thinking

Beginning in the 1950s, the DCAD theory brought fresh insights into milk fever prevention. Researchers such as Ender [18], Dishington [19], and later Block [3] found that balancing dietary cations (e.g., potassium, sodium) and anions (e.g., chloride, sulfur) influences calcium metabolism, protecting cows from hypocalcemia near calving. More specifically, feeding a diet with a negative DCAD value promotes calcium mobilization and supports better serum calcium concentrations in the transition period. Subsequently, Horst et al. [20] highlighted that high dietary potassium could impede calcium absorption, rendering dairy cows more vulnerable to milk fever. They provided the first direct evidence that feeding cows a diet lower in potassium decreased the incidence of milk fever. These discoveries propelled a more comprehensive view of disease management by acknowledging the interplay of dietary factors, metabolic processes, and calcium regulation [20]. More recent research has shown that positive DCAD diets improved the inflammatory state as indicated by lower LPS in milk and higher LPS in urine as well as lower LBP and SAA in the serum of cows fed positive DCAD [21].

2.1.4. The Endotoxin Hypothesis and Systemic Inflammation: A New Perspective

Within the last three decades, the endotoxin hypothesis has garnered attention as a compelling explanation for milk fever’s etiology, proposing that Gram-negative bacterial LPS absorbed from mucosal sites (e.g., rumen, udder, or uterus) disrupts calcium homeostasis through systemic inflammation. Aiumlamai et al. [22] initially suggested that endotoxin translocation might aggravate parturient paresis, while Andersen [23] argued that ruminal acidosis allows LPS to breach the rumen barrier, subsequently contributing to laminitis, abomasal displacement, and other production-related diseases. Although the precise mechanisms remain elusive, Ametaj et al. [24,25] advanced the notion that endotoxins may initiate or worsen milk fever and fatty liver, later extending it to multiple periparturient conditions—ranging from ruminal acidosis to retained placenta, mastitis, and downer cow syndrome [10,11,26]. By attributing significant importance to immune-system activation, this new perspective emphasizes that milk fever involves more than just a metabolic issue.

2.1.5. Endotoxins and Systemic Inflammation

Endotoxins strongly stimulate the innate immune response through Toll-like receptor 4 (TLR4), initiating pathways involving nuclear factor-kappa B (NF-κB) and leading to proinflammatory cytokine secretion (tumor necrosis factor (TNF)-α, interleukin (IL)-6, IL-1) [27]. These events manifest as the acute-phase response (APR), which includes fever, acute-phase proteins, and shifts in plasma ion concentrations [28,29]. Augustine et al. [30] reported that TNF-α suppresses parathyroid hormone (PTH) synthesis and inhibits renal 1a-hydroxylase activity, leading to lower levels of 1,25-dihydroxyvitamin D, thereby compromising calcium regulation. Additionally, endotoxins can modify the expression of renal and intestinal Ca2+ transporters [31]. Meanwhile, albumin, lactate, non-esterified fatty acids (NEFA), and certain anions (Cl, SO42−) bind or chelate calcium (Figure 1 and Figure 2), while cells sequester it, collectively exacerbating hypocalcemia just when cows require calcium most. In this context, proinflammatory cytokines further impede bone resorption and calcium mobilization [32]. I am referring to these interconnected metabolic and immune pathways as part of a broader concept of the Calci-Inflammatory Network that sets the stage for Section 4’s in-depth discussion.

3. Inflammatory Response in Milk Fever Cows

A significant feature of milk fever is the presence of a low-grade or subclinical inflammatory state that both precedes and coincides with clinical hypocalcemia. Elevated concentrations of acute-phase proteins [e.g., haptoglobin, serum amyloid A (SAA)] and proinflammatory cytokines (e.g., TNF-α) indicate that systemic inflammation may be integral to milk fever pathogenesis [26,33]. This aligns with the general trend in periparturient disorders, where immune dysregulation and metabolic strain interact.
Ametaj and collaborators further explored this immune component in various periparturient diseases, including milk fever [9,34]. Their findings propose that the inflammatory response mounted against endotoxins may act as both a pathogenic contributor to hypocalcemia and, as a protective mechanism aiming to contain inflammation during the transition period. Such an immune-metabolic synergy offers a more subtle understanding than the traditional “calcium deficiency” paradigm alone. The interplay between inflammation and ionized calcium—which Ametaj is proposing to call the Calci-Inflammatory Network or Axis—anticipates the broader re-evaluation of milk fever theories in Section 4.

4. Re-Evaluating Traditional Theories in Light of the Endotoxin Hypothesis: The Calci-Inflammatory Network

The history of milk fever research demonstrates an evolution from early, often speculative frameworks toward more integrative models. While the 1950 review by Hibbs [12] anchored milk fever in a predominantly metabolic context, subsequent progress with the hypocalcemia theory and the DCAD/potassium hypothesis substantially reduced disease incidence by highlighting dietary and endocrine regulation of calcium [5,6,7,8]. Yet, these metabolic-centered paradigms do not fully account for systemic inflammation and immune activation—especially as emphasized by the endotoxin hypothesis, which proposes that Gram-negative bacterial LPS can trigger an inflammatory cascade that disrupts calcium homeostasis [9,10,11,22,23].
Several publications [10,11] have integrated these findings, leading to the proposal of the Calci-Inflammatory Network (Figure 1), which is a system of interconnected nodes linking inflammation and calcium regulation in periparturient dairy cows. This network encompasses functional interactions between the mammary gland, uterus, rumen, hoof, and parathyroid gland, along with the suppression of intestinal and renal calcium absorption, impaired bone calcium mobilization (generating hypocalcemia), and modulation of immune cells such as neutrophils and macrophages. These interconnected disturbances contribute to the development of periparturient disorders, including mastitis, metritis, and laminitis. At the core of this network is the observation that endotoxin-induced inflammation can lower blood calcium levels, both by (1) altering hormone signaling (e.g., PTH, 1,25-dihydroxyvitamin D) and (2) sequestering Ca2+ into tissues or binding it through acute-phase reactants like albumin or fatty acids. Simultaneously, changes in serum iCa2+—particularly hypocalcemia—can reduce certain proinflammatory signaling cascades, sometimes acting as a physiological brake on overactive immune response [35,36].

4.1. Multiple Sources of Endotoxin and Early Immune Activation

Evidence suggests that LPS or endotoxin can translocate from a range of sites, including the rumen (especially postpartum)—particularly under subacute ruminal acidosis (SARA) conditions [37]—as well as from mastitic udders (both prepartum and postpartum), and metritic uteri, or infected hooves (mainly postpartum) [22,23]. In all these cases, local Gram-negative bacterial growth and tissue damage facilitate LPS leakage into the bloodstream, where it engages Toll-like receptor 4 (TLR4) on immune cells, triggering the release of potent cytokines and acute-phase proteins such as TNF-α, IL-6, and IL-1 and SAA [38,39]. Notably, innate immune activation can be present before the clinical onset of milk fever (i.e., prepartum). Zhang et al. [26] reported increased TNF-α, IL-6, and SAA levels in cows during the dry-off period, well prior to parturition and overt symptoms of milk fever. These findings indicate that a low-grade inflammatory state—potentially arising from subclinical infections and endotoxin translocation—may prime the immune system in advance of calving, setting the stage for subsequent hypocalcemia once lactational calcium demands surge.

4.2. Mechanisms of the Calci-Inflammatory Network

From an immunophysiological standpoint, macrophages and neutrophils depend critically on Ca2+-dependent signaling pathways when activated by LPS (Figure 1). High intracellular Ca2+ levels facilitate the activation of various downstream pathways that drive the release of pro-inflammatory cytokines, including TNF-α, IL-1, and IL-6 [40,41]. This robust cytokine response is crucial for an effective defense against pathogens; however, it can also contribute to tissue damage if left unchecked.
A controlled, transient reduction in intracellular Ca2+—as observed in the periparturient hypocalcemia of cows—appears to serve as a regulatory brake on this inflammatory cascade. By diminishing Ca2+-dependent signaling, the immune cells produce lower quantities of cytokines, thereby potentially mitigating tissue damage that would arise from an overly aggressive inflammatory response [42]. In essence, this temporary hypocalcemic state may act as a natural anti-inflammatory mechanism, preserving tissue integrity during periods of high metabolic and immunological stress.
The Calci-Inflammatory Network, however, can become dysregulated under conditions of chronic or repeated exposure to LPS. In scenarios such as persistent subacute ruminal acidosis (SARA), mastitis, metritis, or untreated hoof infections, the immune system may remain in a prolonged state of activation. This sustained inflammatory state leads to continuous cytokine production, which in turn interferes with calcium homeostasis by antagonizing PTH actions or by altering vitamin D metabolism. As a result, the restoration of normal calcium levels is delayed, perpetuating a cycle where chronic hypocalcemia further undermines neuromuscular function and metabolic stability, while persistent inflammation continues to disrupt calcium mobilization [43].
Adding to this complexity, the CaSR plays a pivotal role in mediating these interactions (Figure 1). Expressed on both the parathyroid glands and various immune cells, CaSR modulates immune responses based on extracellular Ca2+ concentrations. During inflammatory stress, cytokines can downregulate CaSR function, effectively shifting the calcium homeostasis set point and exacerbating hypocalcemia [35,43,44]. Furthermore, the active form of vitamin D, 1,25-dihydroxyvitamin D, may influence both CaSR signaling and immune cell activity, thereby reinforcing the hypocalcemic state under conditions of inflammation [45,46,47].
Collectively, the Calci-Inflammatory Network is not merely a linear feedback loop but a multifaceted network involving receptors, hormones, transport proteins, and cytokines. This network intricately regulates serum calcium levels in response to both physiological and pathological challenges, highlighting the dual role of calcium as both a metabolic substrate and a key modulator of immune function. Understanding this network offers new insights into how transient hypocalcemia may serve as an adaptive response, while also underscoring the risks associated with its chronic dysregulation in dairy cows.

4.3. Clinical Consequences

Seen through this lens, milk fever is neither strictly a metabolic deficiency nor purely an inflammatory disease. Rather, it arises at the intersection of mounting metabolic demands for calcium at parturition and an immune system that may already be hyperreactive. Excessive inflammatory signals impede calcium retrieval from bone and gut, perpetuating hypocalcemia. In turn, prolonged or severe hypocalcemia weakens core physiological functions, increasing the risk of muscle weakness, recumbency, and organ dysfunction. The Calci-Inflammatory Network thus reframes milk fever as a condition wherein systemic inflammation and calcium dysregulation mutually amplify each other’s injurious effects.

4.4. Hypocalcemia in Multiple Periparturient Diseases: The Common Role of Inflammation

Multiple lines of evidence reveal that hypocalcemia is not solely linked to milk fever but frequently occurs in other periparturient diseases, including ketosis, left displaced abomasum (LDA), acute puerperal metritis (APM), and mastitis. In a study by Venjakob et al. [48], cows displaying suboptimal calcium levels around calving showed persistently reduced serum calcium on days 0, 1, and 3 postpartum—an outcome mirrored in these other health conditions. Moreover, ketosis, particularly among first-calf heifers at 3 and 7 days postpartum, consistently features lower calcium concentrations, while cows with LDA are disproportionately prone to developing hypocalcemia [49,50]. Comparable patterns are evident in cows affected by APM, mastitis, retained placenta, dystocia, and left displaced abomasum pointing toward hypocalcemia as both a predisposing factor for these disorders and a by-product of the inflammation they provoke [51,52].
These convergent findings highlight inflammation as a central mechanism linking hypocalcemia with various periparturient diseases. Metabolic disturbances such as ketosis, LDA, APM, and mastitis trigger inflammatory pathways that elevate the release of cytokines like IL-6 and TNF-α, which disrupt calcium equilibrium in several ways. Specifically, cytokines can depress PTH secretion and modify the CaSR, ultimately compromising intestinal calcium absorption, skeletal calcium mobilization, and renal calcium reabsorption [43,53,54]. As inflammation worsens, hypocalcemia intensifies, creating susceptibility to further metabolic or infectious insults and perpetuating a negative feedback loop of immune system impairment and low calcium.
In inflammatory states, the surge in immune-cell activity heightens the need for calcium to fuel fundamental cellular processes. Immunometabolomic research [55] shows that chronic, subclinical inflammation helps sustain this increased calcium requirement. Simultaneously, deepening hypocalcemia weakens neutrophil functionality [56], thereby raising animals’ vulnerability to secondary infections. This two-way interaction magnifies existing metabolic imbalances and promotes a cycle wherein hypocalcemia both stems from and exacerbates ongoing inflammation.
Nutritional factors can compound this cyclical pattern. Conditions like ketosis and mastitis often reduce dry matter intake, limiting dietary calcium precisely when metabolic demands spike. As a consequence, serum calcium decreases even further, impeding the cow’s capability to manage acute-phase responses effectively. Meanwhile, inflammatory mediators impede the usual hormonal processes that restore calcium homeostasis, allowing persistent hypocalcemia to heighten immune dysfunction and metabolic strain.
Central to this cycle is the mutual causation between hypocalcemia and inflammation. Low iCa2+ availability reduces immune-cell efficiency—evidenced by poorer neutrophil performance and diminished immune-cell populations [56]—thereby escalating the odds of infection and subsequent inflammatory events (Figure 1). Conversely, infections and endotoxemia stimulate the production of proinflammatory cytokines that interfere with CaSR and other calcium-regulatory pathways [43]. Studies by Zhang et al. [26] and Abuajamieh et al. [57] further confirm that disordered metabolism and inflammation, acting in unison, are key contributors to conditions like milk fever and ketosis. Such a self-perpetuating feedback loop—where hypocalcemia-induced immunosuppression triggers infection or endotoxin influx, which then reinforces inflammatory activity—emphasizes the necessity of a comprehensive strategy for disease prevention and control.

5. A New Hypothesis on Hypocalcemia and Its Role in Inflammation

Ametaj et al. [10,11] have hypothesized that hypocalcemia, frequently observed during milk fever and also during systemic inflammation and endotoxemia, may serve a protective function by helping control LPS bioactivity in the bloodstream and modulating the systemic inflammatory response. According to this hypothesis, moderate or transient reductions in iCa2+ might limit excessive calcium-dependent activation of immune cells, providing a physiological brake on inflammation [58,59]. Evidence suggests that rather than being an outright deficiency, this hypocalcemia could be a built-in adaptive response aimed at mitigating the harmful effects of endotoxemia and systemic inflammation. An overview of the physiological and pathophysiological ranges of calcium concentrations in plasma and intracellular compartments of periparturient dairy cows is presented in Supplementary Table S1.

5.1. Fate of Endotoxin in the Circulation

Endotoxin (ET), primarily LPS shed in the gastrointestinal tract (GIT) due to high grain or high fat intake, often associates with chylomicrons in epithelial cells, travels via the lymph, and triggers proinflammatory cytokine release by macrophages in the mesenteric lymph node (Figure 2) [60]. Once in circulation, a portion of ET enters the portal vein and is scavenged by immune cells, Kupffer cells, and hepatocytes in the liver, thereby reducing its systemic levels [61]. However, some ET may bypass hepatic clearance and reach systemic circulation, where it binds to high-density lipoprotein (HDL) and low-density lipoprotein (LDL). This binding is mediated by lipopolysaccharide-binding protein (LBP) and soluble CD14 (sCD14) [62,63,64].

5.2. Calcium’s Role in LPS Aggregation and Immune Activation

Ionized calcium influences how LPS behaves in the bloodstream (Figure 2). Under in vitro conditions, divalent cations such as Ca2+ help LPS adopt tightly packed “lamellar” or “multilamellar” structures, generally lowering LPS’s capacity to trigger Toll-like receptor (TLR)-mediated inflammation [65]. Nevertheless, LPS can also occur in various aggregate states, and binding to LBP can rearrange or partially disaggregate LPS [66,67]. When LPS effectively engages macrophages via membrane-bound CD14 (mCD14), a robust cytokine response ensues (e.g., TNF-α, IL-1, IL-6), which is vital for combating infection but detrimental if overactivated [67]. In vivo, elevated intracellular calcium in immune cells can fuel the inflammatory cascade [36,68], potentially aggravating organ damage. Thus, although Ca2+ in plasma can stabilize less-inflammatory LPS structures, excessive Ca2+-dependent signaling inside macrophages or other immune cells may exacerbate inflammation.

5.3. Hypocalcemia as a Protective Mechanism

Ametaj postulates that hypocalcemia, observed during sepsis and systemic inflammation, may serve as an adaptive mechanism to reduce overactivation of the immune system. By lowering iCa2+, the stimulus for calcium-dependent intracellular pathways in macrophages and neutrophils is diminished, limiting the explosive release of proinflammatory cytokines [11]. Indeed, studies show that interventions that reduce iCa2+—for instance, through calcium chelation—can curb LPS-induced mortality, hinting that transient hypocalcemia buffers against excessive endotoxemia [36,69]. Rather than simply aggregating LPS into harmless multilamellar structures, this protective mechanism primarily diminishes the immune-cell hyperactivation that otherwise leads to systemic inflammation and potential organ failure.

5.4. Lipoproteins and LPS Clearance

Regardless of LPS’s structural form (aggregated or partially disaggregated), HDL and LDL serve as major vehicles for LPS clearance (Figure 2). During systemic inflammation, normal HDL (n-HDL) undergoes compositional changes, with SAA replacing apolipoprotein A-I (apoA-I) and forming acute-phase HDL (ap-HDL) [63,64]. Acute-phase HDL exhibits a higher affinity for LPS, promoting its neutralization and hepatic excretion via bile [62,70]. This pathway not only removes LPS from circulation but also prevents excessive immune activation, underscoring the importance of lipoproteins in managing systemic inflammation. In the context of hypocalcemia, dampened immune-cell activity gives lipoprotein-mediated clearance an extended window to operate without provoking large cytokine surges. Thus, the protective advantage of lower iCa2+ may lie in controlling macrophage overactivation and forcing LPS into multilamellar forms.

5.5. Modulation of the Inflammatory Response

By containing calcium-dependent signaling in immune cells and enhancing LPS clearance via lipoproteins, mild hypocalcemia can help modulate the inflammatory response, mitigating the overproduction of cytokines. This is especially relevant in conditions such as endotoxemia and sepsis, where an excessive immune response can be detrimental [64,71]. Indeed, multiple studies in humans and animal models report that elevating iCa2+ during sepsis may worsen outcomes, whereas interventions that keep iCa2+ at lower thresholds can improve survival and reduce organ damage [36]. These findings align with the idea that transient hypocalcemia might function as a physiological safeguard, preventing excessive inflammation until endotoxins are effectively cleared by hepatic macrophages or lipoproteins.

5.6. Interactions of Calcium with Lactate, Fatty Acids, and Albumin

The hypothesis also acknowledges that elevated lactate and NEFA levels, along with reduced albumin concentrations—commonly observed during inflammatory states—can further modulate iCa2+ (Figure 2). These metabolites and proteins bind or chelate ionized calcium, lowering free iCa2+ in the bloodstream. Such binding helps reduce or increase the pool of biologically active calcium (and thus calcium-dependent cellular processes), offering another route by which systemic inflammation might be restricted [72,73,74].

5.7. Lactate Binding to Ionized Calcium

Lactate is produced by anaerobic glycolysis and circulates as the lactate anion (C3H5O3) [72]. It can weakly bind iCa2+ through electrostatic interactions, forming reversible calcium-lactate complexes [73]. Elevated lactate levels in sepsis or lactic acidosis thus transiently lower free iCa2+, potentially affecting nerve function, muscle contraction, blood clotting, and immune-cell activities. The formation of calcium-lactate complexes may help explain the clinical signs of hypocalcemia that appear during severe inflammation or high lactate production [74].

5.8. Non-Esterified Fatty Acids Binding to Ionized Calcium

Non-esterified fatty acids circulate bound to albumin, and their free carboxyl group (-COOH) can also chelate iCa2+ (Figure 2) [75]. The binding affinity can vary with fatty-acid chain length and degree of saturation [75,76]. During metabolic stress—such as the periparturient period in dairy cows—elevated NEFA levels can further reduce free iCa2+, potentially contributing to hypocalcemia’s clinical manifestations [77]. This decline in iCa2+ may help limit some inflammatory processes, although chronic or severe hypocalcemia remains a health risk.

5.9. Albumin Binding to Ionized Calcium

Albumin is the most abundant plasma protein and a key buffer for calcium, as roughly 40–50% of total calcium in the blood is protein-bound (Figure 3) [78]. Negatively charged sites on albumin (carboxyl groups, histidine residues) bind Ca2+, forming a calcium-albumin complex [79]. Although traditionally considered a negative acute-phase protein, albumin can sometimes increase in certain inflammatory contexts, enhancing its calcium-binding capacity and thereby reducing free iCa2+ [80]. This reduction in biologically active calcium may assist in downregulating processes that depend on Ca2+ for inflammatory signaling. Conversely, conditions such as hypoalbuminemia (during acute phase response) increase the fraction of free iCa2+ [81], with potential ramifications for immune-cell activity. Consequently, accurate assessment of calcium status necessitates measuring iCa2+ directly or applying albumin correction to total calcium measurements [82].
Overall, emerging data and the Ametaj et al. [10,11] hypothesis propose that hypocalcemia—commonly observed during sepsis, endotoxemia, or the periparturient period—can act as a physiological modulator of inflammation. While adequate Ca2+ levels can promote lamellar LPS aggregates that are inherently less inflammatory, excessive Ca2+ within immune cells can intensify the inflammatory cascade. By lowering iCa2+, hypocalcemia tempers calcium-dependent immune pathways and allows lipoprotein-mediated clearance of LPS to proceed without triggering widespread cytokine surges (Figure 2). Meanwhile, factors like lactate, NEFAs, and albumin further shape iCa2+ concentrations, linking metabolic and immune homeostasis. This balanced view of calcium’s dual roles—both in LPS structure and in intracellular signaling—helps clarify why a transient decrease in iCa2+ may be protective, even though severe or prolonged hypocalcemia poses its own risks.

6. Calcium Buffering and Sequestration: Distinct Mechanisms and Key Locations in Calcium Homeostasis During Inflammation

Understanding the distinction between calcium buffering and calcium sequestration is essential for grasping calcium homeostasis, particularly in complex physiological states such as systemic inflammation and milk fever in dairy cows. Although these processes are interrelated, they fulfill different roles in regulating the body’s calcium levels. Calcium buffering involves the short-term control of free iCa2+ concentrations to prevent rapid fluctuations that could disrupt cellular functions [83]. In contrast, calcium sequestration refers to the storage of calcium ions in specific cellular compartments or structural reservoirs for long-term management and controlled release [84,85]. This section examines both processes and highlights the main anatomical locations involved, emphasizing their importance in maintaining overall calcium balance—especially under inflammatory conditions.

6.1. Calcium Buffering

Calcium buffering relates to the immediate regulation of free calcium ions within biological fluids and cells. By binding free calcium to proteins and other molecules, buffering stabilizes cytosolic and extracellular iCa2+ levels, protecting crucial physiological processes such as muscle contraction, nerve impulse transmission, and blood clotting. Key binding proteins in this system include albumin in the bloodstream and calmodulin within cells; they help keep free calcium concentrations within a narrow optimal range [86]. Two Supplementary Tables (Tables S1 and S3) are provided, presenting the concentration ranges of iCa2+.

6.1.1. Primary Sites of Calcium Buffering

Blood Plasma

In the bloodstream, around 40% of total calcium is bound to plasma proteins, mostly albumin and globulins (Figure 3). This protein-bound fraction acts as a buffer, ensuring stable free calcium levels needed for physiological functions [78]. By binding calcium ions, these proteins prevent significant swings in iCa2+, the bioactive form of calcium essential for cardiac function and neural transmission [79].

Extracellular Matrix (ECM)

The extracellular matrix (ECM), which comprises proteins like collagen, elastin, and specialized glycoproteins, also participates in buffering extracellular calcium (Figure 3). This buffering is especially important in bone, where the ECM promotes mineralization and contributes to systemic calcium homeostasis [32]. Hydroxyapatite crystals—formed from calcium and phosphate—reside within the collagenous bone matrix, offering both structural integrity and a ready source of calcium. In other tissues, specific ECM molecules (e.g., osteopontin, vitronectin, fibronectin) can bind calcium to stabilize extracellular levels and ensure proper cellular signaling [87].

Intracellular Compartments

Within cells, calcium buffering involves calmodulin in the cytosol and storage in organelles such as the endoplasmic reticulum (ER) and sarcoplasmic reticulum (SR) (Figure 4) [88]. Cytosolic calmodulin binds free calcium, regulating key signaling pathways for muscle contraction, secretion, and metabolism. Meanwhile, the ER and SR temporarily retain calcium, releasing it when signals (e.g., action potentials) demand. This controlled release underlies vital processes such as muscle contraction and neurotransmitter exocytosis [89]. By preventing large shifts in intracellular calcium, this buffering system maintains cellular homeostasis.

6.2. Calcium Sequestration

Calcium sequestration involves storing calcium ions in specialized compartments or structural sites for long-term management [90]. This ensures an accessible supply of calcium during times of high demand, such as growth, pregnancy, lactation, or tissue repair, while preventing unnecessary surges in serum calcium. Major sites of sequestration include bones, the liver, the kidneys, and muscles (Figure 3). By allowing the controlled release of stored calcium, sequestration mechanisms help maintain systemic balance, even during inflammation.

6.2.1. Primary Sites of Calcium Sequestration

Bones

Bones serve as the principal reservoir of calcium in the body, housing roughly 99% of total calcium in the form of hydroxyapatite within the mineralized matrix (Figure 3) [91]. In response to low serum calcium, bone tissue releases calcium to preserve levels necessary for muscle contraction, nerve signaling, and blood coagulation [32]. Parathyroid hormone and calcitonin tightly regulate this process: PTH stimulates osteoclast activity to free calcium under hypocalcemia, whereas calcitonin inhibits osteoclasts to favor deposition during hypercalcemia [92]. This remodeling cycle sustains skeletal integrity while dynamically buffering systemic calcium levels [93]. In dairy cows, persistent inflammatory or metabolic stresses, like those in the periparturient period, can destabilize this remodeling, increasing the likelihood of disorders such as osteoporosis or hypocalcemia [94].

Skeletal Muscles (Sarcoplasmic Reticulum)

In skeletal muscle cells, the SR functions as a critical calcium reservoir (Figure 3). It stores large amounts of calcium ions and releases them into the cytosol when muscles receive electrical signals [95]. The liberated calcium binds troponin, initiating the interaction of actin and myosin for muscle contraction. Subsequently, a SERCA pump drives calcium back into the SR, reducing cytosolic calcium and inducing relaxation [96]. Proper cycling of calcium release and uptake maintains effective muscle function and prevents fatigue [97,98].

Liver (Hepatocytes)

In the liver, hepatocytes store calcium within the ER and mitochondria (Figure 3). This storage supports key intracellular signaling pathways necessary for metabolic processes such as protein synthesis and lipid metabolism [99,100]. Mitochondrial calcium buffering supports ATP generation, matching energy supply to cellular demands. Moreover, calcium-dependent signaling drives hepatic detoxification enzyme activity [101]. Consequently, disturbed calcium handling in hepatocytes, whether via inflammation or other stressors, can disrupt metabolic balance and compromise overall health [102]. By functioning as a major intracellular reservoir, the liver complements bone and kidney roles in systemic calcium regulation [103].

Kidneys (Renal Tubular Cells)

The kidneys maintain calcium equilibrium by controlling its reabsorption or excretion in the renal tubules (Figure 3). Renal tubular cells sequester calcium to conserve it under low-calcium conditions or excrete it to prevent hypercalcemia. Hormones, especially PTH and active vitamin D, modulate these processes, altering calcium reabsorption rates based on systemic needs [104]. Elevated PTH levels, for instance, enhance calcium reabsorption in the distal tubules, minimizing urinary loss during hypocalcemia, whereas vitamin D amplifies tubular cell expression of calcium transporters [104]. When renal calcium handling weakens, imbalances such as chronic hypocalcemia or hypercalcemia can emerge [43].

Brain (Neurons and Glial Cells)

Within the nervous system, neurons and glial cells coordinate calcium sequestration to sustain cellular homeostasis and support synaptic activity (Figure 3). Calcium is primarily stored in the ER and mitochondria, enabling precise regulation of numerous neural processes [105]. In neurons, calcium entering synaptic terminals upon depolarization prompts neurotransmitter release, a crucial step in synaptic plasticity and signal transmission [106]. Additionally, intracellular calcium acts as a second messenger in pathways modulating gene expression, metabolism, and longer-term synaptic adaptations. In glial cells (e.g., astrocytes, microglia), calcium signaling influences neuronal excitability, synaptic strength, and neuroprotection [107]. By buffering excess calcium, glial cells avert excitotoxicity and preserve ionic stability, safeguarding neurons from overstimulation. This intricate neuron-glia interplay is essential for normal brain function, learning, memory, and a healthy neural environment.

Mitochondria

Mitochondria fulfill a dual role in calcium sequestration by combining their well-known function as cellular powerhouses with calcium buffering (Figure 3). Mitochondrial calcium uptake involves the mitochondrial calcium uniporter (MCU), which mediates cytosolic calcium entry into the organelle [108]. Once inside, calcium ions stimulate key citric acid cycle enzymes—such as isocitrate dehydrogenase and α-ketoglutarate dehydrogenase—boosting ATP production to meet the cell’s energy requirements [109]. At the same time, mitochondria sequester excess calcium to prevent cytosolic overload and potential cytotoxic effects [110]. During inflammatory episodes (e.g., mastitis or systemic inflammatory response in dairy cows), cytokines like IL-1 and TNF-α can disrupt normal calcium homeostasis, raising cytosolic calcium and heightening the burden on mitochondria [111]. Excessive mitochondrial calcium uptake under prolonged inflammation may impair ATP synthesis and induce oxidative stress, ultimately exacerbating cellular damage. By balancing energy output and calcium buffering, mitochondria play a pivotal role in shielding cells from inflammatory harm and preserving overall homeostasis.

7. Impact of Systemic Inflammation on Calcium Buffering and Sequestration

Systemic inflammation during the periparturient period in dairy cows significantly affects calcium homeostasis, a critical concern given the high metabolic demands of lactation. Proinflammatory mediators have been shown to impair calcium buffering and sequestration in major organs such as the liver, kidneys, and bone. These mediators disrupt key intracellular structures like the ER and mitochondria [112,113], thereby limiting the cow’s ability to maintain adequate intracellular Ca2+ levels. This impairment increases the risk of hypocalcemia and related clinical issues, including muscle weakness, reduced appetite, and decreased milk production [8,43].
In the liver, cytokines such as IL-1 and TNF-α compromise calcium balance by reducing the ER’s buffering capacity, which is one of the principal calcium reservoirs, and by lowering mitochondrial calcium uptake [114]. These alterations lead to a cytosolic calcium overload, potentially resulting in cellular dysfunction. Systemically, the hepatic disturbances exacerbate hypocalcemia by impairing the liver’s normal role in stabilizing serum calcium levels, thereby aggravating metabolic imbalances [115].
Inflammation also adversely affects renal calcium handling. In the kidneys, particularly within renal tubular cells, inflammation alters the expression and function of calcium transport proteins, such as transient receptor potential (TRP) channels and various calcium ATPases [116]. These changes impair renal reabsorption of calcium, causing increased urinary losses. The resulting reduction in serum calcium is especially problematic during early lactation when calcium demand is elevated, deepening hypocalcemia and increasing the risk of periparturient disorders [117].
Elevated inflammatory cytokines also impact bone metabolism. Inflammatory mediators stimulate osteoclast activity, leading to accelerated bone resorption and the release of stored calcium [118]. Although this process is an attempt to maintain serum calcium levels, the excessive periparturient demands can overwhelm the bone’s ability to release sufficient calcium. Persistent inflammation ultimately disrupts the balance of bone remodeling, weakening the skeletal structure while failing to meet systemic calcium requirements [119].
Skeletal muscles, particularly through the action of the SR, also suffer under inflammatory stress. In skeletal muscle cells, inflammation can impair calcium handling by the SR, as cytokine-driven increases in calcium demand for immune responses reduce the available pool for muscle contraction [120]. This mechanism contributes to muscle weakness and recumbency, classical signs of severe hypocalcemia during clinical milk fever. Additionally, oxidative stress induced by inflammation may degrade the SERCA pump, diminishing its ability to re-sequester calcium into the SR [121].
Platelet function is not exempt from the effects of systemic inflammation. Inflammatory mediators can modulate calcium dynamics within platelets; high intracellular calcium levels can intensify platelet activation and clot formation, which serves as a protective mechanism against hemorrhage but also increases the risk of thrombotic complications under chronic inflammatory conditions [122]. Conversely, hypocalcemia can hamper platelet function and delay clot formation, thereby complicating hemostasis and healing outcomes [123].
Red blood cells (RBCs) and immune cells are similarly affected by systemic inflammation. In RBCs, excess intracellular calcium reduces cell deformability, thereby shortening their lifespan and undermining effective oxygen delivery to tissues [124].
In immune cells, calcium plays a dual regulatory role in inflammation. On one hand, hypocalcemia impairs cytokine production by limiting calcium-dependent intracellular signaling cascades in immune cells. On the other hand, hypercalcemia—via activation of the calcium-sensing receptor (CaSR)—can amplify inflammatory responses by promoting cytokine release and inflammasome activation. Thus, calcium homeostasis is tightly linked to the modulation of immune responses, and disruptions in either direction can influence the trajectory of inflammatory diseases [125].
The brain is also impacted by these inflammatory processes. Neurons and glial cells experience disrupted calcium buffering under systemic inflammation, contributing to excitotoxicity and neuroinflammatory processes [126]. Such alterations can compromise central regulatory mechanisms for calcium homeostasis, thereby compounding hypocalcemia and impairing the overall physiological response [127].
Vascular endothelial cells are particularly sensitive to inflammatory signals, which provoke increased calcium influx and impair their ability to sequester calcium effectively. This imbalance can alter vascular tone and permeability [128], leading to excessive vasodilation, promotion of leukocyte adhesion, and ultimately vascular instability. Concurrently, low circulating Ca2+ levels further worsen perfusion deficits and compromise end-organ function [80].
Inflammation also triggers the remodeling of the extracellular matrix (ECM), which can release bound calcium into the circulation [129]. Although this release may briefly boost serum calcium levels, the resulting fluctuations can degrade tissue integrity, promote local injury, and contribute to maladaptive remodeling, such as fibrosis [130].
In vascular smooth muscle cells, inflammation-induced dysregulation of calcium homeostasis can impair blood pressure control [131]. Cytokine-driven modifications of calcium handling in these cells can lead to either hypertension or hypotension, both of which jeopardize tissue perfusion and exacerbate inflammatory complications [132].
The mechanisms underlying hypocalcemia during systemic inflammation are multifactorial. Cytokine-driven changes in calcium handling and tissue-level responses promote intracellular sequestration and binding, reducing free serum calcium. While these processes may serve to protect tissues from calcium overload, the net reduction in available Ca2+ diminishes normal cellular functions and further intensifies inflammation and clinical pathology [43,133]. Additionally, inflammatory states drive the formation of insoluble calcium complexes with plasma anions such as lactate, chloride, and phosphate, which further reduces the availability of free Ca2+ [134]. Although this reduction can help curb excessive inflammatory signaling, it also compromises critical processes such as muscle contraction and immune function, thereby complicating disease management [135].
Finally, various cell types actively sequester intracellular Ca2+ during inflammation to prevent cytotoxicity. Endothelial cells, immune cells, and hepatocytes utilize pumps and transporters—including SERCA, PMCA, MCU, and NCX—to govern intracellular calcium flux. Proper functioning of these transport systems is essential for regulating immune responses and maintaining tissue integrity, thereby preventing a detrimental cycle of sustained inflammation and impaired calcium homeostasis [136].

8. Intracellular Calcium Buffers: Pathways and Components Involved in Calcium Homeostasis

Intracellular calcium is a pivotal second messenger that regulates numerous cellular processes, and its concentration is tightly controlled by an elaborate network of buffering, sequestration, and transport mechanisms (Figure 4). The SR in muscle cells and ER in non-muscle cells serve as the primary reservoir for Ca2+, orchestrating its rapid sequestration and release during muscle contraction and relaxation. Key calcium-binding proteins such as calsequestrin, sarcalumenin, junctate, and triadin facilitate these processes. For instance, calsequestrin binds Ca2+ with high capacity, enabling the storage of large quantities of calcium that can be quickly mobilized when required [137,138]. Proteins like sarcalumenin and junctate further modulate calcium dynamics, while triadin is critical for anchoring these proteins to the ryanodine receptor complex, thereby optimizing excitation-contraction coupling [139].
In the cytoplasm, soluble calcium-binding proteins such as calmodulin, calbindin-D28K, calbindin-D9K, and calreticulin transiently bind free Ca2+, serving as dynamic buffers that modulate both the amplitude and duration of calcium signals. Calmodulin, in particular, functions as a versatile calcium sensor, interacting with a broad range of enzymes, ion channels, and other proteins to influence diverse processes from metabolism to gene transcription [140,141]. This buffering capacity is crucial for ensuring that cellular calcium levels remain within a narrow range optimal for proper cell function.
Membrane-associated proteins also play an essential role in maintaining intracellular calcium homeostasis (Figure 4). The sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) pump is responsible for actively transporting Ca2+ from the cytoplasm back into the SR/ER, utilizing ATP to re-establish basal cytosolic calcium levels following cellular signaling events [142]. Additionally, transient receptor potential (TRP) channels facilitate Ca2+ entry in response to a variety of stimuli [143], while the sodium-calcium exchanger (NCX) helps extrude Ca2+ from cells by exchanging it for sodium ions, thereby contributing to the overall balance of intracellular calcium [144].
The plasma membrane Ca2+-ATPase (PMCA) is a critical membrane protein responsible for the active extrusion of Ca2+ from the cytosol, thereby maintaining low intracellular calcium levels. Utilizing ATP, PMCA rapidly decreases cytosolic Ca2+ following cellular activation, which is essential for terminating calcium signals. Multiple isoforms of PMCA, each with distinct regulatory properties, allow fine-tuning of calcium homeostasis in various tissues [145]. Disruptions in PMCA function have been linked to several pathophysiological conditions, highlighting its vital role in cellular health.
Beyond these classical compartments, lysosomes have emerged as significant secondary Ca2+ stores. Calcium exchange between lysosomes and the ER occurs through two-pore calcium channels (TPC1 and TPC2), which enable localized calcium signaling events crucial for processes such as autophagy and vesicular trafficking [146]. Furthermore, the ER-mitochondria junctions serve as specialized sites for efficient calcium transfer. At these junctions, inositol trisphosphate receptors (IP3Rs) on the ER interface with voltage-dependent anion channels (VDACs) on mitochondria, facilitating the transfer of Ca2+ into mitochondria via the rapid uptake mode (RaM). This process is essential not only for buffering cytosolic Ca2+ but also for supporting key metabolic functions, including ATP production and the regulation of apoptotic pathways [147].
Another critical aspect of calcium homeostasis involves the ER-plasma membrane junction, where store-operated calcium entry (SOCE) replenishes ER calcium stores. SOCE is mediated by the interaction between ORAI channels and stromal interaction molecule 1 (STIM1). When ER calcium levels fall, STIM1 undergoes a conformational change, aggregates near the plasma membrane, and activates ORAI channels to permit Ca2+ influx, thereby restoring calcium balance within the ER [148]. The Golgi apparatus further contributes to intracellular calcium buffering by serving as a calcium reservoir that is vital for vesicular trafficking and protein modification. Calcium within the Golgi plays an important role in the sorting and maturation of secretory proteins, thereby aiding in the spatial and temporal regulation of calcium signaling within the cell [149].
Together, these interconnected systems highlight the complexity of intracellular Ca2+ regulation. The coordinated activity of the SR/ER, cytoplasmic buffers, membrane transport proteins, lysosomal channels, and inter-organelle communication ensures that cells can respond precisely to physiological stimuli while maintaining calcium homeostasis. A deep understanding of these pathways not only enhances our grasp of fundamental cellular physiology but also holds significant implications for therapeutic strategies in conditions where calcium regulation is disrupted.

9. The Calcium-Sensing Receptor: Functions, Mechanisms, and Role in Immune Modulation

The calcium-sensing receptor is a G-protein-coupled receptor (GPCR) originally discovered in the parathyroid gland but is now recognized as a widely expressed molecule with pivotal roles in calcium homeostasis, tissue physiology, and immune regulation [150,151]. Structurally, the CaSR is characterized by a large extracellular Venus flytrap domain, a seven-transmembrane domain, and an intracellular C-terminal region that interacts with multiple signaling proteins, including G proteins and β-arrestins [152,153]. Through these components, CaSR integrates diverse extracellular stimuli to initiate downstream intracellular responses, including the modulation of cyclic AMP (cAMP), mitogen-activated protein kinase (MAPK) cascades, phospholipase C (PLC)-inositol 1,4,5-trisphosphate (IP3) signaling, and intracellular calcium fluxes [154].

9.1. CaSR Function in Calcium Homeostasis

The CaSR serves as the central sensor of extracellular calcium concentrations [Ca2+]e and regulates systemic calcium balance through its actions in the parathyroid gland, kidneys, intestine, and bone [154,155]. Upon increased [Ca2+]e, CaSR activation suppresses parathyroid hormone (PTH) secretion, reduces renal calcium reabsorption, inhibits the synthesis of 1,25-dihydroxyvitamin D3, and decreases intestinal calcium absorption. Inflammatory conditions can upregulate CaSR expression or enhance its sensitivity, particularly in the parathyroid gland and kidneys. This heightened calcium sensing leads to inappropriate suppression of parathyroid hormone (PTH), increased urinary calcium excretion, and reduced intestinal calcium absorption due to lower calcitriol levels—all of which contribute to the development or worsening of hypocalcemia [156].
The mechanism by which the CaSR suppresses PTH secretion is related to its abundant expression on parathyroid chief cells suppressing PTH secretion in response to elevated [Ca2+]e concentrations. Upon binding calcium, CaSR activates intracellular signaling cascades via G-proteins, particularly Gq/11 and Gi/o. The Gq/11 pathway stimulates phospholipase C, leading to inositol trisphosphate (IP3)-mediated calcium release from intracellular stores and activation of protein kinase C (PKC), both of which inhibit PTH exocytosis. Simultaneously, the Gi/o pathway reduces cyclic AMP (cAMP) levels, thereby suppressing PTH gene transcription. These pathways converge to downregulate PTH synthesis and secretion, ensuring tight regulation of systemic calcium levels [150,151,156].

9.2. CaSR in Immune Regulation and Inflammation

The CaSR is also expressed in various immune cells, including monocytes, macrophages, neutrophils, dendritic cells, and lymphocytes [157]. In these cells, CaSR activation modulates multiple immune functions, notably cytokine secretion, chemotaxis, phagocytosis, and inflammasome activation. Studies have shown that extracellular calcium acts as a chemotactic agent for macrophages via CaSR and that CaSR-mediated signaling enhances the secretion of IL-1β and IL-6 through activation of the NF-κB and MAPK pathways [158,159]. Furthermore, CaSR activation in monocytes and macrophages contributes to NLRP3 inflammasome activation and promotes antigen presentation via constitutive macropinocytosis [160,161]. In neutrophils, CaSR modulates activation, oxidative burst, and cytokine production through NF-κB [162]. In T lymphocytes, CaSR facilitates cytokine secretion, T-cell apoptosis, and possibly memory cell formation via PLC-IP3 and TRPC channels [163,164]. Dysregulation of CaSR in these immune compartments has been associated with pathologies including sepsis, myocardial infarction, rheumatoid arthritis, and autoimmune orchitis [165,166,167]. The role of CaSR in dairy cows remains poorly understood, highlighting a critical need for further research in this area.

9.3. Hypocalcemia, Calcium Sequestration, and Immune Cell Impairment

During inflammation, systemic hypocalcemia is often observed due to increased CaSR expression and inflammatory cytokine activity (e.g., IL-1β, IL-6, TNF-α) [168]. At the cellular level, calcium is sequestered into the ER and mitochondria to prevent cytotoxicity and support ATP production. However, this sequestration can reduce cytosolic calcium availability, impairing calcium-dependent signaling in immune cells. Neutrophils show impaired chemotaxis, phagocytosis, and ROS production due to blunted PLC-IP3 signaling [46]. Macrophages display reduced phagocytic capacity, cytokine output, and M1 polarization, along with suppressed NFAT and NF-κB activation [169]. T cells exhibit impaired activation and proliferation via reduced calcineurin-NFAT signaling and altered Th subset differentiation [170]. B cells experience diminished antibody production due to disrupted BCR signaling [171]. Dendritic cells show impaired antigen presentation and IL-12 secretion, leading to weakened adaptive immunity [172].

9.4. Sequestration Benefits and Risks

Calcium buffering in the endoplasmic ER and mitochondria protects against calcium overload and supports cellular metabolism [147,173]. However, during prolonged inflammation or systemic hypocalcemia, persistent sequestration depletes cytosolic calcium needed for immune activation [89,174]. This leads to immunosuppression and reduced pathogen clearance, especially in the context of periparturient diseases in dairy cows and critical illness in humans [58,67]. The CaSR is a multifunctional receptor that integrates extracellular calcium-sensing with endocrine, metabolic, and immune signaling [150,152]. While essential for maintaining calcium homeostasis, its upregulation during inflammation contributes to hypocalcemia and immune modulation [159]. Calcium sequestration, although protective, can impair immune cell function under prolonged stress or deficiency. Understanding CaSR-mediated signaling pathways in immune cells opens avenues for therapeutic interventions in inflammation, sepsis, metabolic disorders, and calcium-related diseases [151,153].

10. Systems Integration of Calcium Buffers and Inflammatory Networks

The preceding sections detailed the individual roles of albumin, high-density lipoproteins (HDL), apolipoproteins, and inorganic anions in modulating calcium availability and inflammation. In this section, we move beyond isolated mechanisms to present a cohesive systems-level framework, wherein calcium buffering and immune regulation are interconnected through molecular feedback loops. This integrated view positions these buffers as dynamic mediators in the Calci-Inflammatory Network, coordinating mineral metabolism and host defense.

10.1. Cross-Talk Between Calcium Buffering and Inflammatory Signaling

Under physiological conditions, albumin, HDL, and inorganic anions maintain ionic homeostasis, transport metabolites, and contribute to the neutralization of bacterial components. During inflammation, however, their functionality becomes deeply intertwined with immune activation. Hypoalbuminemia, acute-phase HDL remodeling and metabolic acidosis collectively reshape the ionized calcium (iCa2+) pool, which in turn modulates cytokine release and leukocyte behavior.
Albumin, the primary carrier of calcium in plasma, binds up to 45% of total circulating calcium. Its concentration declines during the acute phase response, reducing total calcium levels while transiently increasing the free iCa2+ fraction. However, inflammatory mediators and associated metabolites (e.g., lactate, NEFA) can chelate calcium, driving a net hypocalcemic effect. These shifts affect intracellular calcium signaling in immune cells, altering their threshold for activation and cytokine release [175,176,177].

10.2. HDL and Apolipoproteins: Facilitators of Endotoxin Clearance

HDL and its primary protein component, apolipoprotein A-I (ApoA-I), serve as first-line detoxifiers of lipopolysaccharide (LPS), a potent endotoxin derived from Gram-negative bacteria. During inflammation, HDL undergoes acute-phase remodeling into apHDL, in which ApoA-I is replaced by serum amyloid A (SAA). While apHDL may exhibit reduced cholesterol efflux and antioxidant functions, its capacity to neutralize and eliminate LPS is markedly enhanced.
This remodeling shortens HDL’s circulatory half-life from approximately 4 days to 2–4 h, promoting rapid hepatic clearance of LPS through scavenger receptors (e.g., Stabilin-1/2) on liver sinusoidal endothelial cells [178,179,180]. In parallel, free ApoA-I, dissociated during HDL remodeling, binds LPS and contributes to its renal excretion, complementing the hepatic pathway [181,182]. Rather than impairing function, apHDL represents an adaptive reconfiguration that accelerates LPS detoxification and enhances host defense during acute systemic inflammation.

10.3. Inorganic Anions and the pH–Calcium–Inflammation Network

Inorganic anions such as chloride, sulfate, phosphate, and bicarbonate modulate both extracellular pH and calcium dynamics. Through dietary manipulation of the dietary cation-anion difference (DCAD), it is possible to induce a compensated metabolic acidosis that promotes calcium mobilization. This strategy is used to enhance calcium availability in transition dairy cows. However, excessive acidification can enhance oxidative stress and leukocyte activation, thereby linking ion balance to immune activation [183,184].
Anionic shifts not only influence iCa2+ levels via changes in albumin binding and calcium solubility but also contribute to an inflammatory phenotype when chronic or unregulated. Thus, inorganic anions function as metabolic modulators that integrate nutritional status, mineral metabolism, and immune tone.

10.4. Emergent Network Properties and Translational Potential

When considered collectively, these buffers exhibit emergent properties that go beyond their individual actions. They participate in a network of feedback loops—regulated by cytokines (IL-1β, IL-6, TNF-α), hormones (PTH, cortisol, vitamin D), and metabolic cues—that maintain or disrupt calcium homeostasis during inflammatory stress [185,186,187].
Clinically, this integrated perspective offers new avenues for intervention. Simultaneous measurement of iCa2+, albumin, and HDL functionality may serve as a composite biomarker of mineral and immune balance. Nutritional manipulation of DCAD or therapeutic modulation of apHDL remodeling could be leveraged to accelerate recovery from systemic inflammation. Recognizing these molecular buffers as part of a unified Calci-Inflammatory Network opens novel possibilities for diagnostics and precision management in both veterinary and human medicine.

11. Evidence Linking Hypocalcemia to Inflammatory Responses in Dairy Cows

11.1. Recent Research in Dairy Cows Supporting the Hypothesis

Recent studies provide substantial evidence supporting the hypothesis that hypocalcemia serves as an adaptive response to modulate inflammation during endotoxemia. This evidence emphasizes the protective role of hypocalcemia in controlling systemic inflammation and offers new insights into managing periparturient diseases in dairy cows. For instance, Chandler et al. [183] described significant changes in calcium dynamics following an acute intravenous (iv) LPS challenge in early postpartum cows. Their research showed that serum total calcium (tCa) and iCa concentrations decreased following the LPS challenge, supporting the idea that hypocalcemia is a response to inflammation rather than merely a deficiency state. The decrease in calcium levels was not attributable to urinary excretion, suggesting that other mechanisms, such as calcium sequestration in tissues, contribute to hypocalcemia during inflammatory challenges [183].

11.2. Exacerbation of Inflammation by High Calcium Levels

Additional research has demonstrated that maintaining high calcium levels during immune challenges can exacerbate inflammation, confirming that hypocalcemia helps alleviate the inflammatory response. For example, Horst et al. [184] investigated the effects of maintaining eucalcemia in dairy cows subjected to LPS-induced endotoxemia. They found that cows with elevated calcium levels exhibited significantly increased levels of inflammatory markers, such as LBP and SAA, compared to cows with hypocalcemia. This study highlights the aggravation of systemic inflammation due to high calcium levels, supporting the hypothesis that hypocalcemia serves as a protective mechanism [147].

11.3. Experimental Designs Inducing Endotoxemia

Various experimental designs, including the administration of LPS to induce endotoxemia, have consistently shown significant decreases in serum calcium concentrations following immune activation. This decline in calcium levels appears to be dose-dependent and temporally correlated with the inflammatory response. In a study conducted by Waldron et al. [185] different doses of LPS were administered to lactating cows, leading to a dose-dependent decrease in serum calcium levels. The findings indicate that immune activation via LPS infusion disrupts calcium homeostasis, thereby underscoring the role of hypocalcemia in controlling inflammation [185].

11.4. Hypocalcemia as a Response to Endotoxin-Induced Systemic Inflammation

In the experiment conducted by Horst et al. [184], they aimed to evaluate the effects of maintaining eucalcemia on metabolic, immune, and production variables during an LPS challenge in lactating dairy cows. The study involved two groups: cows receiving LPS alone (LPS-Con) and cows receiving LPS along with intravenous calcium to maintain eucalcemia (LPS-Ca). Cows in the LPS-Ca group demonstrated heightened inflammatory responses, with significantly increased levels of acute-phase proteins (LBP and SAA) compared to the LPS-Con group. Maintaining eucalcemia also led to reduced neutrophil counts and an exacerbated febrile response, but neutrophil function remained unaffected.
Interestingly, milk yield decreased more in the LPS-Ca group, and their recovery to baseline dry matter intake (DMI) was delayed, suggesting the negative effects of calcium infusion on production performance. The study further highlighted that hypocalcemia, commonly observed during systemic inflammation, may play a role in regulating immune responses by influencing calcium homeostasis and inflammatory signaling pathways. These findings support the hypothesis that hypocalcemia helps relieve inflammation triggered by intravenous endotoxin infusion, serving as a protective mechanism to prevent excessive immune activation.

11.5. Pre-Existing Systemic Inflammation in Cows Prone to Milk Fever

Research focusing on cows susceptible to milk fever has revealed significant alterations in acute phase proteins and pro-inflammatory cytokines before the onset of the disease and during the disease occurrence. Ametaj et al. [34] and Zhang et al. [26] reported that cows prone to developing milk fever exhibited elevated concentrations of pro-inflammatory cytokines, such as TNF-α and acute phase proteins such as SAA, weeks before parturition as well as at parturition week. These findings suggest a pre-existing state of systemic inflammation in cows prone to milk fever, which persists postpartum. Elevated levels of these inflammatory markers indicate that systemic inflammation is a critical factor in the pathogenesis of milk fever [26,34].
In another study, Seminara et al. [186] provide further evidence that the acute phase response is closely intertwined with the development of hypocalcemia (i.e., low total serum calcium concentrations) in postpartum dairy cows. Their research demonstrated that cows that developed dyscalcemia exhibited significant increases in acute phase proteins such as serum amyloid A (SAA), haptoglobin, and lipopolysaccharide-binding protein (LBP) within the first four days postpartum—an increase not observed in eucalcemic cows. These findings suggest that inflammatory activation is not merely a precursor but may also contribute mechanistically to calcium dysregulation.
Specifically, elevated concentrations of SAA and haptoglobin were temporally associated with the onset of hypocalcemia in dyscalcemic cows, suggesting that systemic inflammation may disrupt calcium homeostasis through effects on calcium-binding proteins, renal excretion, or hormonal signaling. This supports the hypothesis that postpartum inflammation and calcium imbalance are interconnected processes, with inflammation playing a central role in destabilizing calcium metabolism. The study adds to a growing body of evidence supporting a Calci-Inflammatory Network, linking immune activation with metabolic disturbances such as hypocalcemia during the early postpartum period.
These findings underscore the importance of further research to elucidate the molecular mechanisms bridging inflammation and calcium regulation, with the goal of improving strategies to prevent postpartum calcium-related disorders in dairy cows.

11.6. Calcium Oral Supplementation and Its Role in Postpartum Inflammation in Dairy Cows

Couto Serrenho et al. [187] investigated the effects of postpartum calcium (Ca) supplementation on systemic inflammation in clinically healthy multiparous dairy cows. The study involved 101 cows assigned to either receive two oral Ca boluses (TRT) or no supplementation (CON) within 24 h of calving. Blood Ca levels and markers of inflammation, including Hp, SAA, and LBP, were measured over eight days postpartum.
The TRT group exhibited transiently higher total Ca levels immediately after supplementation but lower Ca levels by day 2 compared to controls. Concentrations of SAA were significantly higher in the TRT group on day 2, reflecting increased systemic inflammation. Similarly, younger multiparous cows (parity 2) in the TRT group showed higher LBP levels, suggesting heightened inflammatory responses compared to older cows.
Haptoglobin and albumin levels, however, were unaffected by treatment, indicating a selective inflammatory response. These results suggest that supplemental Ca may modestly increase markers of inflammation postpartum, particularly in cows less predisposed to hypocalcemia. The study also highlights the potential for Ca supplementation to disrupt natural postpartum calcium homeostasis.

12. Translational Implications of the Calci-Inflammatory Network

The Calci-Inflammatory Network, originally conceptualized in the context of periparturient hypocalcemia in dairy cows, provides a unifying physiological and pathological framework that extends far beyond veterinary medicine. This integrative model reveals conserved biological interactions between calcium homeostasis and inflammatory responses, which appear to operate similarly across species—including in humans.
In human medicine, hypocalcemia is a frequent and well-documented feature in critically ill patients, particularly in conditions such as sepsis, systemic inflammatory response syndrome (SIRS), acute pancreatitis, and trauma. Numerous studies have shown that decreased ionized calcium (iCa2+) levels in these patients are strongly associated with increased disease severity and mortality [175,188,189]. These human syndromes mirror key pathophysiological features of milk fever in dairy cows, including elevated proinflammatory cytokines (e.g., TNF-α, IL-6), hypoalbuminemia, elevated lactate and NEFA levels, and decreased iCa2+ availability.
Importantly, some human studies suggest that rapid intravenous correction of hypocalcemia may paradoxically worsen inflammation, potentially due to abrupt intracellular calcium loading that promotes oxidative stress, mitochondrial dysfunction, or cell death [133,175]. These findings align with the central hypothesis of the Calci-Inflammatory Network—that transient hypocalcemia may represent an adaptive, immunomodulatory response during acute systemic stress, rather than a purely pathological deficiency.
In veterinary medicine, calcium therapy for clinical milk fever is a well-established practice. However, recent findings by McArt and Neves [189] underscore the complexity of postpartum hypocalcemia by identifying distinct phenotypes—transient, persistent, and delayed. These data suggest that a one-size-fits-all approach to calcium supplementation may be suboptimal and that precision calcium therapy, aligned with the cow’s individual inflammatory and metabolic profile, is warranted. Overcorrection of calcium levels may overstimulate calcium-sensing receptors (CaSR) on immune cells, potentially exacerbating inflammatory signaling.
Moreover, the Calci-Inflammatory Network supports ongoing efforts to identify biomarkers and regulatory feedback loops that integrate inflammation with calcium metabolism. Examples include reduced renal 1α-hydroxylase activity (leading to impaired activation of vitamin D), increased calcium binding to acute phase reactants (e.g., albumin, NEFA, lactate), and CaSR-mediated immune cell signaling, all of which have been documented in both bovine and human studies [10,175,190].
Ultimately, the Calci-Inflammatory Network calls for a paradigm shift: hypocalcemia during inflammatory stress should be considered potentially physiological and protective, rather than strictly pathological. This redefinition opens new avenues for research into therapeutic modulation of calcium–inflammation crosstalk, early diagnostic tools using metabolomics, and the development of immunometabolic therapies that aim to fine-tune, rather than override, the body’s natural homeostatic responses.

13. The Role of Calcium in LPS Aggregation and Clearance Without Inducing Inflammation

Lipopolysaccharide, a major component of Gram-negative bacterial outer membranes, can trigger potent inflammation via the LBP–CD14–TLR4 axis. Its bioactivity is strongly dictated by its structural conformation [191,192]. Under many in vitro and in vivo conditions, divalent cations—particularly Ca2+ and Ba2+—promote the transition of LPS from “fluid” or “micellar” aggregate phases into multilamellar aggregates [193]. These lamellar forms exhibit reduced fluidity, diminished acyl-chain mobility, and tighter lipid packing, making them less accessible to LBP or CD14 [193]. As a result, multilamellar LPS is less capable of activating TLR4, which translates into reduced production of pro-inflammatory cytokines [194]. Such calcium-induced LPS aggregates are generally recognized as relatively inert particles or “debris” that Kupffer cells and other macrophages can phagocytose without prompting a substantial inflammatory burst [195,196].
The clearance of these aggregated LPS particles proceeds via multiple non-inflammatory routes. Scavenger receptors (e.g., SR-A1, SR-B1) on macrophages readily bind the structurally modified or aggregated endotoxins, promoting their endocytosis and subsequent lysosomal degradation [197,198]. Additionally, plasma lipoproteins and proteins such as HDL and lactoferrin bind lamellar LPS, further reducing its capacity to stimulate TLR4 and directing it toward hepatic clearance [199,200]. Ultimately, Kupffer cells degrade LPS internally and excrete the fragments into bile, from where they are eliminated through the gastrointestinal tract (Figure 2) [198]. In some instances, LPS–calcium complexes may be temporarily stored in adipose tissue, where tissue-resident macrophages gradually remove them [201,202]. Because the multilamellar conformation is less immunogenic, these clearance processes remain largely non-inflammatory [203].
Together, these findings help reconcile the dual roles of calcium. On one hand, elevated extracellular Ca2+ promotes the formation of lamellar LPS aggregates that neutralize its toxicity. On the other, transient hypocalcemia in vivo can be advantageous by restraining calcium-dependent pro-inflammatory signaling within immune cells [35,192]. Rather than being contradictory, these phenomena—lamellar neutralization of LPS in the presence of adequate Ca2+ and immune diminishing under moderate hypocalcemia—can coexist, each providing protective mechanisms at different phases of the inflammatory response or in distinct physiological compartments. This view emphasizes the delicate balance required: calcium must be available in sufficient amounts to stabilize and inactivate LPS structurally, yet excessive calcium-driven immune stimulation can be harmful if it promotes enhanced cytokine release. Further studies focusing on these dual roles of calcium in LPS conformation and immune-cell activation are needed to optimize preventive and therapeutic strategies for endotoxemia, sepsis, and periparturient diseases in dairy cattle [204].

14. Conclusions

This review advances a new conceptual framework for understanding hypocalcemia in milk fever, challenging the long-standing notion that calcium fluctuations are merely pathological deficiencies requiring urgent correction. Instead, we propose that changes in calcium levels represent adaptive, dynamic responses that participate in the regulation of systemic inflammation. At the core of this framework is the Calci-Inflammatory Network, which highlights how oscillations in calcium—modulated in part by the calcium-sensing receptor (CaSR) on immune cells, as well as in parathyroid and renal tissues—fulfill a dual regulatory role.
On one hand, elevated intracellular calcium supports antimicrobial defense mechanisms, including the neutralization of LPS and the activation of calcium-dependent immune pathways. On the other hand, transient hypocalcemia appears to function as a physiological brake, tempering immune hyperactivation, reducing cytokine overproduction, and minimizing collateral tissue damage and organ dysfunction.
Multiple studies, including those by Chandler et al. [183], Horst et al. [184], Waldron et al. [185], Seminara et al. [186], Couto-Serrenho et al. [187], Ametaj [34], and Zhang [26], provide compelling evidence in support of this model. Collectively, these findings underscore that calcium fluctuations are not random nor deleterious per se, but rather essential for maintaining a delicate balance between effective host defense and the avoidance of a hyperinflammatory state. Furthermore, CaSR-mediated feedback in parathyroid and renal tissues contributes to the fine-tuning of parathyroid hormone (PTH) and 1,25-dihydroxyvitamin D production, reinforcing the integration between calcium regulation and immune modulation.
The implications of this emerging paradigm extend beyond traditional milk fever management. It calls for a precision-based approach to calcium therapy—one that aligns calcium supplementation with the beneficial immunomodulatory effects of controlled calcium fluctuations. Future intervention strategies should aim not merely to normalize calcium levels, but to optimize them in concert with immune function, thereby enhancing overall health and resilience during the critical periparturient period.
Further research into the genetic, environmental, and nutritional determinants of calcium dynamics will be pivotal for developing targeted, evidence-based protocols. By integrating this knowledge into herd management, the dairy industry stands to improve animal welfare, disease prevention, productivity, and long-term sustainability.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/dairy6030022/s1. Table S1. Physiological and Pathophysiological Concentrations of Calcium in Plasma and Intracellular Compartments in Dairy Cows Around Parturition. This table summarizes the ranges of total and ionized calcium in plasma, interstitial fluid, and intracellular compartments under normal and disease states. It provides comparative benchmarks for interpreting calcium dysregulation in clinical and subclinical milk fever, highlighting homeorhetic versus pathological adaptations. Table S2. Plasma Endotoxin Concentrations in Cattle and Human Subjects. This table compiles data from multiple studies on circulating endotoxin levels (EU/mL or pg/mL) in both healthy and diseased states in cattle and humans. It includes values under conditions such as subacute ruminal acidosis, displaced abomasum, ketosis, and critical illness in humans, offering insights into endotoxemia across species. Table S3. Serum Total Calcium and Ionized Calcium Thresholds in Dairy Cows. This table presents threshold values that define normocalcemia, subclinical hypocalcemia, and clinical hypocalcemia in dairy cows, with reference to both total and ionized calcium concentrations. It serves as a clinical classification tool for the diagnosis and monitoring of calcium-related disorders in the periparturient period.

Funding

This review article received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

I would like to express my gratitude to Zohaib Saleem for his invaluable assistance in developing the figures for this review article. I deeply appreciate his diligence, skill, and persistence in mastering the BioRender software, which he used to create all the figures included in this article. His contributions have significantly enhanced the quality and clarity of the article.

Conflicts of Interest

The author declares no conflicts of interest.

References

  1. Goff, J.P. The monitoring, prevention, and treatment of milk fever and subclinical hypocalcemia in dairy cows. Vet. J. 2008, 176, 50–57. [Google Scholar] [CrossRef] [PubMed]
  2. Reinhardt, T.A.; Lippolis, J.D.; McCluskey, B.J.; Goff, J.P.; Horst, R.L. Prevalence of subclinical hypocalcemia in dairy herds. J. Dairy Sci. 2011, 94, 2928–2934. [Google Scholar] [CrossRef]
  3. Block, E. Manipulating dietary anions and cations for prepartum dairy cows to reduce incidence of milk fever. J. Dairy Sci. 1984, 67, 2939–2948. [Google Scholar] [CrossRef] [PubMed]
  4. Lean, I.J.; DeGaris, P.J.; McNeil, D.M.; Block, E. Hypocalcemia in dairy cows: Pathophysiology and preventive strategies. J. Dairy Sci. 2006, 89, 1094–1108. [Google Scholar] [CrossRef]
  5. Block, E. Manipulation of dietary cation-anion difference on nutritionally related production diseases, productivity, and metabolic responses of dairy cows. J. Dairy Sci. 1994, 77, 1437–1450. [Google Scholar] [CrossRef]
  6. Venjakob, P.L.; Borchardt, S.; Heuwieser, W. Hypocalcemia—Cow-level prevalence and preventive strategies in German dairy herds. J. Dairy Sci. 2017, 100, 9258–9266. [Google Scholar] [CrossRef]
  7. DeGaris, P.J.; Lean, I.J. Milk fever in dairy cows: A review of pathophysiology and control principles. Vet. J. 2008, 176, 58–69. [Google Scholar] [CrossRef]
  8. Serrenho, R.C.; DeVries, T.J.; Duffield, T.F.; LeBlanc, S.J. Graduate student literature review: What do we know about the effects of clinical and subclinical hypocalcemia on health and performance of dairy cows? J. Dairy Sci. 2021, 104, 6304–6326. [Google Scholar] [CrossRef]
  9. Zebeli, Q.; Beitz, D.C.; Bradford, B.J.; Dunn, S.M.; Ametaj, B.N. Peripartal alterations of calcitonin gene-related peptide and minerals in dairy cows affected by milk fever. Vet. Clin. Pathol. 2013, 42, 70–77. [Google Scholar] [CrossRef]
  10. Ametaj, B.N.; Zebeli, Q.; Iqbal, S. Nutrition, microbiota, and endotoxin-related diseases in dairy cows. Rev. Bras. Zootec. 2010, 39, 433–444. [Google Scholar] [CrossRef]
  11. Eckel, E.F.; Ametaj, B.N. Invited review: Role of bacterial endotoxins in the etiopathogenesis of periparturient diseases of transition dairy cows. J. Dairy Sci. 2016, 99, 5967–5990. [Google Scholar] [CrossRef] [PubMed]
  12. Hibbs, J.W. Milk fever (Parturient paresis) in dairy cows: A review. J. Dairy Sci. 1950, 33, 758–789. [Google Scholar] [CrossRef]
  13. Hutyra, F.; Marek, J.; Manniger, R. Special Pathology and Therapeutics of the Diseases of Domestic Animals, 4th ed.; Greig, J.R., Ed.; Forgotten Books: London, UK, 1938. [Google Scholar]
  14. Dryerre, H.; Greig, J.R. Milk fever: Its possible association with derangements in the internal secretions. Vet. Rec. 1925, 5, 225–231. [Google Scholar]
  15. Auger, M.L. Réalisation expérimentale de la fièvre vitulaire. Compt. Rend. 1926, 182, 348–350. [Google Scholar]
  16. Little, W.L.; Wright, N.C. The aetiology of milk fever in cattle. Br. J. Exp. Pathol. 1925, 6, 129. [Google Scholar]
  17. Dryrre, H.; Greig, J.R. The specific chemotherapy of milk fever by the parenteral administration of Ca-boro-gluconate. Vet. Med. 1935, 30, 234–238. [Google Scholar]
  18. Ender, F.; Dishington, I.W.; Helgebostad, A. Calcium balance studies in dairy cows under experimental induction and prevention of hypocalcaemic paresis puerperalis. The solution of the aetiology and the prevention of milk fever by dietary means. Z. Tierphysiol. Tierernaehr. Futtermittelkd. 1971, 28, 233. [Google Scholar]
  19. Dishington, I.W. Prevention of milk fever (Hypocalcaemia paresis puerperalis) by dietary salt supplements. Acta Vet. Scand. 1975, 16, 503–512. [Google Scholar] [CrossRef]
  20. Horst, R.L.; Goff, J.P.; Reinhardt, T.A.; Buxton, D.R. Strategies for preventing milk fever in dairy cattle. J. Dairy Sci. 1997, 80, 1269–1280. [Google Scholar] [CrossRef]
  21. Wang, Z.; Li, X.Y.; Yu, Y.N.; Yang, L.Y.; Zhang, P.H.; He, J.H.; Shen, W.J.; Tan, Z.L.; Feng, B.L.; Tang, S.X. Enhancing dietary cation-anion difference reshaped the distribution of endotoxin across different biofluids and influenced inflammatory response in dairy cows exposed to heat stress. Anim. Feed. Sci. Technol. 2020, 262, 114444. [Google Scholar] [CrossRef]
  22. Aiumlamai, S.; Kindahl, H.; Fredriksson, G.; Edqvist, L.E.; Kulander, L.; Eriksson, O. The role of endotoxins in induced ruminal acidosis in calves. Acta Vet. Scand. 1992, 33, 117–127. [Google Scholar] [CrossRef] [PubMed]
  23. Andersen, P.H. Bovine endotoxicosis: Some aspects of relevance to production diseases. A review. Acta Vet. Scand. Suppl. 2003, 98, 141–155. [Google Scholar] [CrossRef]
  24. Ametaj, B.N. A new understanding of the causes of fatty liver in dairy cows. Adv. Dairy Technol. 2005, 17, 97–112. [Google Scholar]
  25. Ametaj, B.N. Strong relationships between mediators of the acute phase response and fatty liver in dairy cows. Can. J. Dairy Sci. 2005, 85, 165–175. [Google Scholar] [CrossRef]
  26. Zhang, G.; Dervishi, E.; Ametaj, B.N. Milk fever in dairy cows is preceded by activation of innate immunity and alterations in carbohydrate metabolism prior to disease occurrence. Res. Vet. Sci. 2018, 117, 167–177. [Google Scholar] [CrossRef]
  27. Shandilya, U.; Sharma, A.; Mallikarjunappa, S.; Guo, J.; Mao, Y.; Meade, K.; Karrow, N. CRISPR-Cas9-mediated knockout of TLR4 modulates Mycobacterium avium ssp. paratuberculosis cell lysate-induced inflammation in bovine mammary epithelial cells. J. Dairy Sci. 2021, 104, 11135–11146. [Google Scholar] [CrossRef] [PubMed]
  28. Carroll, J.; Reuter, R.; Chase, C.; Coleman, S.; Riley, D.; Spiers, D.; Arthington, J.; Galyean, M. Profile of the bovine acute-phase response following an intravenous bolus-dose lipopolysaccharide challenge. Innate Immun. 2009, 15, 81–89. [Google Scholar] [CrossRef]
  29. Jermann, P.; Wagner, L.; Fritsche, D.; Gross, J.; Wellnitz, O.; Bruckmaier, R. Acute phase reaction to LPS-induced mastitis in early lactation dairy cows fed nitrogenic, glucogenic or lipogenic diets. J. Dairy Sci. 2023, 106, 9879–9891. [Google Scholar] [CrossRef] [PubMed]
  30. Augustine, M.; Leonard, M.; Thayu, M.; Baldassano, R.; De Boer, I.; Shults, J.; Denson, L.; DeBoer, M.; Herskovitz, R.; Denburg, M. Changes in vitamin D-related mineral metabolism after induction with anti-tumor necrosis factor-α therapy in Crohn’s disease. J. Clin. Endocrinol. Metab. 2014, 99, E991–E998. [Google Scholar] [CrossRef]
  31. Meurer, M.; Höcherl, K. Endotoxaemia differentially regulates the expression of renal Ca2+ transport proteins in mice. Acta Physiol. 2018, 225, e13175. [Google Scholar] [CrossRef]
  32. Klein, G. The role of calcium in inflammation-associated bone resorption. Biomolecules 2018, 8, 69. [Google Scholar] [CrossRef] [PubMed]
  33. Dervishi, E.; Ametaj, B.N. Milk fever: Reductionist versus systems veterinary approach. In Periparturient Diseases of Dairy Cows; Ametaj, B.N., Ed.; Springer: Berlin, Germany, 2017; pp. 247–266. [Google Scholar]
  34. Ametaj, B.N.; Goff, J.P.; Horst, R.L.; Bradford, B.J.; Beitz, D.C. Presence of acute phase response in normal and milk fever dairy cows around parturition. Acta Vet. Scand. Suppl. 2003, 98, 241. [Google Scholar] [CrossRef]
  35. Thaveeratitham, P.; Khovidhunkit, W.; Patumraj, S. High-density lipoproteins (HDL) inhibit endotoxin-induced leukocyte adhesion on endothelial cells in rats: Effect of the acute-phase HDL. Clin. Hemorheol. Microcirc. 2007, 36, 1–12. [Google Scholar] [PubMed]
  36. Collage, R.D.; Howell, G.M.; Zhang, X.; Stripay, J.L.; Lee, J.S.; Angus, D.C.; Rosengart, M.R. Calcium supplementation during sepsis exacerbates organ failure and mortality via calcium/calmodulin-dependent protein kinase kinase signaling. Crit. Care Med. 2013, 41, e352–e360. [Google Scholar] [CrossRef] [PubMed]
  37. Emmanuel, D.G.; Madsen, K.L.; Churchill, T.A.; Dunn, S.M.; Ametaj, B.N. Acidosis and lipopolysaccharide from Escherichia coli B:055 cause hyperpermeability of rumen and colon tissues. J. Dairy Sci. 2007, 90, 5552–5557. [Google Scholar] [CrossRef]
  38. Su, G.; Klein, R.; Aminlari, A.; Zhang, H.; Steinstraesser, L.; Alarcon, W.; Remick, D.; Wang, S. Kupffer cell activation by lipopolysaccharide in rats: Role for lipopolysaccharide binding protein and toll-like receptor 4. Hepatology 2000, 31, 932–936. [Google Scholar] [CrossRef]
  39. Hu, X.; Li, S.; Mu, R.; Guo, J.; Zhao, C.; Cao, Y.; Zhang, N.; Fu, Y. The rumen microbiota contributes to the development of mastitis in dairy cows. Microbiol. Spectr. 2022, 10, e0251221. [Google Scholar] [CrossRef]
  40. Papaioannou, N.; Voutsas, I.; Samara, P.; Tsitsilonis, O. A flow cytometric approach for studying alterations in the cytoplasmic concentration of calcium ions in immune cells following stimulation with thymic peptides. Cell. Immunol. 2016, 302, 32–40. [Google Scholar] [CrossRef]
  41. Yang, R.; Mark, M.; Gray, A.; Huang, A.; Xie, M.; Zhang, M.; Goddard, A.; Wood, W.; Gurney, A.; Godowski, P. Toll-like receptor-2 mediates lipopolysaccharide-induced cellular signalling. Nature 1998, 395, 284–288. [Google Scholar] [CrossRef]
  42. Iamartino, L.; Brandi, M. The calcium-sensing receptor in inflammation: Recent updates. Front. Physiol. 2022, 13, 1059369. [Google Scholar] [CrossRef]
  43. Hendy, G.; Canaff, L. Calcium-sensing receptor, proinflammatory cytokines and calcium homeostasis. Semin. Cell Dev. Biol. 2016, 49, 37–43. [Google Scholar] [CrossRef] [PubMed]
  44. Brown, E.M. Role of the calcium-sensing receptor in extracellular calcium homeostasis. Nat. Rev. Endocrinol. 2013, 9, 391–403. [Google Scholar] [CrossRef]
  45. Kehrli, M.E., Jr.; Goff, J.P. Periparturient hypocalcemia in cows: Effects on peripheral blood neutrophil and lymphocyte function. J. Dairy Sci. 1989, 72, 1188–1196. [Google Scholar] [CrossRef] [PubMed]
  46. Kimura, K.; Reinhardt, T.A.; Goff, J.P. Parturition and hypocalcemia blunts calcium signals in immune cells of dairy cattle. J. Dairy Sci. 2006, 89, 2588–2595. [Google Scholar] [CrossRef]
  47. Canaff, L.; Hendy, G. Human calcium-sensing receptor gene. J. Biol. Chem. 2002, 277, 30337–30350. [Google Scholar] [CrossRef] [PubMed]
  48. Venjakob, P.; Staufenbiel, R.; Heuwieser, W.; Borchardt, S. Association between serum calcium dynamics around parturition and common postpartum diseases in dairy cows. J. Dairy Sci. 2020, 104, 2243–2253. [Google Scholar] [CrossRef]
  49. Venjakob, P.; Staufenbiel, R.; Heuwieser, W.; Borchardt, S. Serum calcium dynamics within the first 3 days in milk and the associated risk of acute puerperal metritis. J. Dairy Sci. 2019, 102, 11428–11438. [Google Scholar] [CrossRef]
  50. Rodríguez, E.M.; Arís, A.; Bach, A. Associations between subclinical hypocalcemia and postparturient diseases in dairy cows. J. Dairy Sci. 2017, 100, 7427–7434. [Google Scholar] [CrossRef]
  51. Atalay, H. The effect of serum β-hydroxybutyric acid and calcium levels on left displaced abomasum in Holstein cows on transition period. J. Istanbul Vet. Sci. 2019, 3, 43–48. [Google Scholar] [CrossRef]
  52. Curtis, C.; Erb, H.; Sniffen, C.; Smith, R.; Powers, P.; Smith, M.; Me, W.; Rb, H.; Pearson, E. Association of parturient hypocalcemia with eight periparturient disorders in Holstein cows. J. Am. Vet. Med. Assoc. 1983, 183, 559–561. [Google Scholar] [CrossRef]
  53. Kantham, L.; Quinn, S.; Egbuna, O.; Baxi, K.; Butters, R.; Pang, J.; Pollak, M.; Goltzman, D.; Brown, E. The calcium-sensing receptor (CaSR) defends against hypercalcemia independently of its regulation of parathyroid hormone secretion. Am. J. Physiol. Endocrinol. Metab. 2009, 297, E915–E923. [Google Scholar] [CrossRef]
  54. Marmalyuk, D.; Runova, G.; Fadeyev, V. The role of the calcium-sensing receptor in the regulation of parathyroid hormone secretion in physiology and in calcitropic diseases. Osteoporos. Bone Dis. 2023, 26, 25–32. [Google Scholar] [CrossRef]
  55. Zhang, B.X.; Huang, B.; Jiang, Q.; Loor, J.; Lv, X.; Zhang, W.; Li, M.; Wen, J.; Yin, Y.; Wang, J.; et al. Transcriptomics of circulating neutrophils in dairy cows with subclinical hypocalcemia. Front. Vet. Sci. 2022, 9, 959831. [Google Scholar] [CrossRef]
  56. Martinez, N.; Sinedino, L.; Bisinotto, R.; Ribeiro, E.; Gomes, G.; Lima, F.; Greco, L.; Risco, C.; Galvão, K.; Taylor-Rodriguez, D.; et al. Effect of induced subclinical hypocalcemia on physiological responses and neutrophil function in dairy cows. J. Dairy Sci. 2014, 97, 874–887. [Google Scholar] [CrossRef] [PubMed]
  57. Abuajamieh, M.; Kvidera, S.K.; Fernandez, M.V.; Nayeri, A.; Upah, N.C.; Nolan, E.A.; Lei, S.M.; DeFrain, J.M.; Green, H.B.; Schoenberg, K.M.; et al. Inflammatory biomarkers are associated with ketosis in periparturient Holstein cows. Res. Vet. Sci. 2016, 109, 81–85. [Google Scholar] [CrossRef]
  58. Kimura, K.; Goff, J.P.; Kehrli, M.E., Jr.; Reinhardt, T.A. Decreased neutrophil function as a cause of retained placenta in dairy cattle. J. Dairy Sci. 2002, 85, 544–550. [Google Scholar] [CrossRef] [PubMed]
  59. Trebak, M.; Kinet, J. Calcium signalling in T cells. Nat. Rev. Immunol. 2019, 19, 154–169. [Google Scholar] [CrossRef] [PubMed]
  60. Ghoshal, S.; Witta, J.; Zhong, J.; de Villiers, W.; Eckhardt, E. Chylomicrons promote intestinal absorption of lipopolysaccharides. J. Lipid Res. 2009, 50, 90–97. [Google Scholar] [CrossRef]
  61. Trebicka, J.; Krag, A.; Gansweid, S.; Appenrodt, B.; Schiedermaier, P.; Sauerbruch, T.; Spengler, U. Endotoxin and tumor necrosis factor-receptor levels in portal and hepatic vein of patients with alcoholic liver cirrhosis receiving elective transjugular intrahepatic portosystemic shunt. Eur. J. Gastroenterol. Hepatol. 2011, 23, 1218–1225. [Google Scholar] [CrossRef]
  62. Munford, R.S.; Hall, C.L.; Dietschy, J.M. Binding of Salmonella typhimurium lipopolysaccharides to rat high-density lipoproteins. Infect. Immun. 1981, 34, 835–843. [Google Scholar] [CrossRef]
  63. Wurfel, M.M.; Wright, S.D. Lipopolysaccharide (LPS) binding protein catalyzes binding of LPS to lipoproteins. Prog. Clin. Biol. Res. 1995, 392, 287–295. [Google Scholar] [PubMed]
  64. Kitchens, R.L.; Wolfbauer, G.; Albers, J.J.; Munford, R.S. Plasma lipoproteins promote the release of bacterial lipopolysaccharide from the monocyte cell surface. J. Biol. Chem. 1999, 274, 34116–34122. [Google Scholar] [CrossRef]
  65. Tobias, P.S.; Soldau, K.; Ulevitch, R.J. Identification of a lipid A binding site in the acute phase reactant lipopolysaccharide binding protein. J. Biol. Chem. 1989, 264, 10867–10871. [Google Scholar] [CrossRef]
  66. Wright, S.D.; Ramos, R.A.; Tobias, P.S.; Ulevitch, R.J.; Mathison, J.C. CD14, a receptor for complexes of lipopolysaccharide (LPS) and LPS binding protein. Science 1990, 249, 1431–1433. [Google Scholar] [CrossRef] [PubMed]
  67. Dobrovolskaia, M.A.; Vogel, S.N. Toll receptors, CD14, and macrophage activation and deactivation by LPS. Microbes Infect. 2002, 4, 903–914. [Google Scholar] [CrossRef] [PubMed]
  68. Zaloga, G.P. Ionized hypocalcemia during sepsis. Crit. Care Med. 2000, 28, 266–268. [Google Scholar] [CrossRef]
  69. Skarnes, R.C.; Chedid, L. Biological degradation and inactivation of endotoxin (chromate-labeled). In Bacterial Endotoxins; Landy, M., Braun, W., Eds.; Rutgers University Press: New Brunswick, NJ, USA, 1964; pp. 575–587. [Google Scholar]
  70. Hotchkiss, R.S.; Karl, I.E. Calcium: A regulator of the inflammatory response in endotoxemia and sepsis. New Horiz. 1996, 4, 58–71. [Google Scholar]
  71. Feingold, K.R.; Grunfeld, C. Lipids: A key player in the battle between the host and microorganisms. J. Lipid Res. 2012, 53, 2487–2489. [Google Scholar] [CrossRef]
  72. Gibot, S. On the origins of lactate during sepsis. Crit. Care 2012, 16, 151. [Google Scholar] [CrossRef]
  73. Toffaletti, J.; Abrams, B. Effects of in vivo and in vitro production of lactic acid on ionized, protein-bound, and complex-bound calcium in blood. Clin. Chem. 1989, 35, 935–938. [Google Scholar] [CrossRef]
  74. Khatua, B.; Yaron, J.R.; El-Kurdi, B.; Kostenko, S.; Papachristou, G.I.; Singh, V.P. Ringer’s lactate prevents early organ failure by providing extracellular calcium. J. Clin. Med. 2020, 9, 263. [Google Scholar] [CrossRef]
  75. Zaloga, G.P.; Willey, S.; Tomasic, P.; Chernow, B. Free fatty acids alter calcium binding: A cause for misinterpretation of serum calcium values and hypocalcemia in critical illness. J. Clin. Endocrinol. Metab. 1987, 64, 1010–1014. [Google Scholar] [CrossRef] [PubMed]
  76. Whitsett, J.; Tsang, R.C. In vitro effects of fatty acids on serum-ionized calcium. J. Pediatr. 1977, 91, 233–236. [Google Scholar] [CrossRef]
  77. Chapinal, N.; Leblanc, S.J.; Carson, M.E.; Leslie, K.E.; Godden, S.; Capel, M.; Santos, J.E.; Overton, M.W.; Duffield, T.F. Herd-level association of serum metabolites in the transition period with disease, milk production, and early lactation reproductive performance. J. Dairy Sci. 2012, 95, 5676–5682. [Google Scholar] [CrossRef] [PubMed]
  78. Pedersen, K.O. Protein-bound calcium in human serum: Quantitative examination of binding and its variables by a molecular binding model and clinical chemical implications for measurement of ionized calcium. Scand. J. Clin. Lab. Investig. 1972, 30, 321–329. [Google Scholar] [CrossRef]
  79. Dimeski, G.; Treacy, O. The influence of albumin and pH on total and ionized calcium and magnesium. Point Care 2018, 17, 123–126. [Google Scholar] [CrossRef]
  80. Dalal, P.; Muller, W.; Sullivan, D. Endothelial cell calcium signaling during barrier function and inflammation. Am. J. Pathol. 2019, 190, 535–542. [Google Scholar] [CrossRef]
  81. Ridefelt, P.; Helmersson-Karlqvist, J. Albumin adjustment of total calcium does not improve the estimation of calcium status. Scand. J. Clin. Lab. Investig. 2017, 77, 442–447. [Google Scholar] [CrossRef]
  82. Buege, M.; Do, B.; Lee, H.; Weber, D.; Horowitz, S.; Feng, L.; Qing, Y.; Shank, B. Corrected calcium versus ionized calcium measurements for identifying hypercalcemia in patients with multiple myeloma. Cancer Treat. Res. Commun. 2019, 21, 100159. [Google Scholar] [CrossRef]
  83. Gilabert, J. Cytoplasmic calcium buffering: An integrative crosstalk. Adv. Exp. Med. Biol. 2020, 1131, 163–182. [Google Scholar] [CrossRef]
  84. Kraus-Friedmann, N. Calcium sequestration in the liver. Cell Calcium 1990, 11, 625–640. [Google Scholar] [CrossRef] [PubMed]
  85. Giorgi, C.; Marchi, S.; Pinton, P. The machineries, regulation and cellular functions of mitochondrial calcium. Nat. Rev. Mol. Cell Biol. 2018, 19, 713–730. [Google Scholar] [CrossRef]
  86. Baird, G.S. Ionized calcium. Clin. Chim. Acta 2011, 412, 696–701. [Google Scholar] [CrossRef] [PubMed]
  87. Contri, M.; Boraldi, F.; Taparelli, F.; Paepe, A.; Ronchetti, I. Matrix proteins with high affinity for calcium ions are associated with mineralization within the elastic fibers of pseudoxanthoma elasticum dermis. Am. J. Pathol. 1996, 148, 569–577. [Google Scholar]
  88. Hoeflich, K.P.; Ikura, M. Calmodulin in action: Diversity in target recognition and activation mechanisms. Cell 2002, 108, 739–742. [Google Scholar] [CrossRef]
  89. Berridge, M.J.; Bootman, M.D.; Roderick, H.L. Calcium signalling: Dynamics, homeostasis and remodelling. Nat. Rev. Mol. Cell Biol. 2003, 4, 517–529. [Google Scholar] [CrossRef] [PubMed]
  90. Bazil, J.N.; Blomeyer, C.A.; Pradhan, R.K.; Camara, A.K.; Dash, R.K. Modeling the calcium sequestration system in isolated guinea pig cardiac mitochondria. J. Bioenerg. Biomembr. 2013, 45, 177–188. [Google Scholar] [CrossRef]
  91. Vannucci, L.; Fossi, C.; Quattrini, S.; Guasti, L.; Pampaloni, B.; Gronchi, G.; Giusti, F.; Romagnoli, C.; Cianferotti, L.; Marcucci, G.; et al. Calcium intake in bone health: A focus on calcium-rich mineral waters. Nutrients 2018, 10, 1930. [Google Scholar] [CrossRef]
  92. Zikan, V.; Stepan, J.J. Marker of bone resorption in acute response to exogenous or endogenous parathyroid hormone. Biomark. Insights 2008, 3, 19–24. [Google Scholar] [CrossRef]
  93. Cianferotti, L.; Gomes, A.R.; Fabbri, S.; Tanini, A.; Brandi, M.L. The calcium-sensing receptor in bone metabolism: From bench to bedside and back. Osteoporos. Int. 2015, 26, 2055–2071. [Google Scholar] [CrossRef]
  94. Hatate, K.; Kawashima, C.; Hanada, M.; Kayano, M.; Yamagishi, N. Short communication: Serum osteoprotegerin concentrations in periparturient dairy cows. J. Dairy Sci. 2018, 101, 6622–6626. [Google Scholar] [CrossRef]
  95. Roux, E.; Marhl, M. Role of sarcoplasmic reticulum and mitochondria in Ca2+ removal in airway myocytes. Biophys. J. 2004, 86, 2583–2595. [Google Scholar] [CrossRef]
  96. Stammers, A.; Susser, S.; Hamm, N.; Hlynsky, M.; Kimber, D.; Kehler, D.; Duhamel, T. The regulation of sarco(endo)plasmic reticulum calcium-ATPases (SERCA). Can. J. Physiol. Pharmacol. 2015, 93, 843–854. [Google Scholar] [CrossRef] [PubMed]
  97. Treves, S.; Jungbluth, H.; Muntoni, F.; Zorzato, F. Congenital muscle disorders with cores: The ryanodine receptor calcium channel paradigm. Curr. Opin. Pharmacol. 2008, 8, 319–326. [Google Scholar] [CrossRef] [PubMed]
  98. Michelucci, A.; García-Castañeda, M.; Boncompagni, S.; Dirksen, R.T. Role of STIM1/ORAI1-mediated store-operated Ca2+ entry in skeletal muscle physiology and disease. Cell Calcium 2018, 76, 101–115. [Google Scholar] [CrossRef] [PubMed]
  99. Barritt, G. Calcium signalling in liver cells. In Calcium Signalling in Liver Cells; Springer: Berlin/Heidelberg, Germany, 2000; pp. 73–94. [Google Scholar] [CrossRef]
  100. Rieusset, J. Endoplasmic reticulum–mitochondria calcium signaling in hepatic metabolic diseases. Biochim. Biophys. Acta Mol. Cell Res. 2017, 1864, 865–876. [Google Scholar] [CrossRef]
  101. Oliva-Vilarnau, N.; Hankeova, S.; Vorrink, S.U.; Mkrtchian, S.; Andersson, E.R.; Lauschke, V.M. Calcium signaling in liver injury and regeneration. Front. Med. 2018, 5, 192. [Google Scholar] [CrossRef]
  102. Jagtap, Y.; Adlakha, N. Simulation of buffered advection diffusion of calcium in a hepatocyte cell. Math. Biol. Bioinform. 2018, 13, 609–619. [Google Scholar] [CrossRef]
  103. Kang, Y.; McKenna, T.; Watson, L.; Williams, R.; Holt, M. Cytochemical changes in hepatocytes of rats with endotoxemia or sepsis: Localization of fibronectin, calcium, and enzymes. J. Histochem. Cytochem. 1988, 36, 665–678. [Google Scholar] [CrossRef]
  104. Bernardo, J.; Friedman, P. Renal calcium metabolism. In Renal Physiology; Academic Press: London, UK, 2013; pp. 2225–2247. [Google Scholar] [CrossRef]
  105. Lim, D.; Semyanov, A.; Genazzani, A.; Verkhratsky, A. Calcium signaling in neuroglia. Int. Rev. Cell Mol. Biol. 2021, 362, 1–53. [Google Scholar] [CrossRef]
  106. Wang, F.; Yuan, T.; Pereira, A.; Verkhratsky, A.; Huang, J. Glial cells and synaptic plasticity. Neural Plast. 2016, 2016, 5042902. [Google Scholar] [CrossRef] [PubMed]
  107. Clementi, E.; Racchetti, G.; Melino, G.; Meldolesi, J. Cytosolic Ca2+ buffering, a cell property that in some neurons markedly decreases during aging, has a protective effect against NMDA/nitric oxide-induced excitotoxicity. Life Sci. 1996, 59, 389–397. [Google Scholar] [CrossRef] [PubMed]
  108. Yamada, A.; Watanabe, A.; Yamamoto, T. Regulatory mechanisms of mitochondrial calcium uptake by the calcium uniporter complex. Biophys. Physicobiol. 2023, 20, e200004. [Google Scholar] [CrossRef] [PubMed]
  109. Denton, R. Regulation of mitochondrial dehydrogenases by calcium ions. Biochim. Biophys. Acta 2009, 1787, 1309–1316. [Google Scholar] [CrossRef]
  110. Alvarez, S.; Evelson, P.; Vanasco, V.; Magnani, N.; Cimolai, M.; Marchini, T. The role of mitochondria in inflammatory syndromes. In Mitochondria in Inflammation; Tech Science Press: Nanjing, China, 2016; pp. 233–247. [Google Scholar] [CrossRef]
  111. Zhang, H.; Kovacs-Nolan, J.; Kodera, T.; Eto, Y.; Mine, Y. γ-Glutamyl cysteine and γ-glutamyl valine inhibit TNF-α signaling in intestinal epithelial cells and reduce inflammation in a mouse model of colitis via allosteric activation of the calcium-sensing receptor. Biochim. Biophys. Acta 2015, 1852, 792–804. [Google Scholar] [CrossRef]
  112. Rieusset, J.; Fauconnier, J.; Paillard, M.; Belaidi, E.; Tubbs, E.; Chauvin, M.; Durand, A.; Bravard, A.; Teixeira, G.; Bartosch, B.; et al. Disruption of calcium transfer from ER to mitochondria links alterations of mitochondria-associated ER membrane integrity to hepatic insulin resistance. Diabetologia 2016, 59, 614–623. [Google Scholar] [CrossRef]
  113. Li, F.; Guan, Z.; Gao, Y.; Bai, Y.; Zhan, X.; Ji, X.; Xu, J.; Zhou, H.; Rao, Z. ER stress promotes mitochondrial calcium overload and activates the ROS/NLRP3 axis to mediate fatty liver ischemic injury. Hepatol. Commun. 2024, 8, e0399. [Google Scholar] [CrossRef]
  114. Briones-Suarez, L.; Cifuentes, M.; Bravo-Sagua, R. Secretory factors from calcium-sensing receptor-activated SW872 pre-adipocytes induce cellular senescence and a mitochondrial fragmentation-mediated inflammatory response in HepG2 cells. Int. J. Mol. Sci. 2023, 24, 5217. [Google Scholar] [CrossRef]
  115. Williams, G.S.; Boyman, L.; Chikando, A.C.; Khairallah, R.J.; Lederer, W.J. Mitochondrial calcium uptake. Proc. Natl. Acad. Sci. USA 2013, 110, 10479–10486. [Google Scholar] [CrossRef]
  116. Zhou, Y.; Greka, A. Calcium-permeable ion channels in the kidney. Am. J. Physiol. Renal Physiol. 2016, 310, F1157–F1167. [Google Scholar] [CrossRef]
  117. Woudenberg-Vrenken, T.; Bindels, R.; Hoenderop, J. The role of transient receptor potential channels in kidney disease. Nat. Rev. Nephrol. 2009, 5, 441–449. [Google Scholar] [CrossRef] [PubMed]
  118. Klein, G.; Castro, S.; Garofalo, R. The calcium-sensing receptor as a mediator of inflammation. Semin. Cell Dev. Biol. 2016, 49, 52–56. [Google Scholar] [CrossRef]
  119. Zhao, B. Does TNF promote or restrain osteoclastogenesis and inflammatory bone resorption? Crit. Rev. Immunol. 2018, 38, 253–261. [Google Scholar] [CrossRef] [PubMed]
  120. Tupling, A. The sarcoplasmic reticulum in muscle fatigue and disease: Role of the sarco(endo)plasmic reticulum Ca2+-ATPase. Can. J. Appl. Physiol. 2004, 29, 308–329. [Google Scholar] [CrossRef] [PubMed]
  121. Qaisar, R.; Bhaskaran, S.; Ranjit, R.; Premkumar, P.; Huseman, K.; Sataranatarajan, K.; Van Remmen, H. Restoration of SERCA ATPase as an intervention to muscle impairment associated with oxidative stress. FASEB J. 2018, 32, 618-15. [Google Scholar] [CrossRef]
  122. Mussbacher, M.; Salzmann, M.; Brostjan, C.; Hoesel, B.; Schoergenhofer, C.; Datler, H.; Hohensinner, P.; Basílio, J.; Petzelbauer, P.; Assinger, A.; et al. Cell type-specific roles of NF-κB linking inflammation and thrombosis. Front. Immunol. 2019, 10, 85. [Google Scholar] [CrossRef]
  123. Esmon, C. The impact of the inflammatory response on coagulation. Thromb. Res. 2004, 114, 321–327. [Google Scholar] [CrossRef]
  124. Mohanty, J.; Barodka, V.; Berkowitz, D.; Rifkind, J. Intracellular calcium mediated stiffness of red blood cells is reversed by hypoxic pre-incubation with nitrite ions. Biophys. J. 2010, 98, 77a. [Google Scholar] [CrossRef]
  125. Lacy, P.; Stow, J. Cytokine release from innate immune cells: Association with diverse membrane trafficking pathways. Blood 2011, 118, 9–18. [Google Scholar] [CrossRef]
  126. Kirdajova, D.; Kriska, J.; Tureckova, J.; Anděrová, M. Ischemia-triggered glutamate excitotoxicity from the perspective of glial cells. Front. Cell. Neurosci. 2020, 14, 51. [Google Scholar] [CrossRef]
  127. Arundine, M.; Tymianski, M. Molecular mechanisms of calcium-dependent neurodegeneration in excitotoxicity. Cell Calcium 2003, 34, 325–337. [Google Scholar] [CrossRef] [PubMed]
  128. Muller, W. How endothelial cells regulate transmigration of leukocytes in the inflammatory response. Am. J. Pathol. 2014, 184, 886–896. [Google Scholar] [CrossRef] [PubMed]
  129. Shannon, T.; Ginsburg, K.; Bers, D. Potentiation of fractional sarcoplasmic reticulum calcium release by total and free intra-sarcoplasmic reticulum calcium concentration. Biophys. J. 2000, 78, 334–343. [Google Scholar] [CrossRef] [PubMed]
  130. Cataldi, A.; Giacomo, V. Extracellular matrix: Immunity and inflammation. In Extracellular Matrix; Springer: Berlin/Heidelberg, Germany, 2018; pp. 83–109. [Google Scholar] [CrossRef]
  131. Touyz, R.; Alves-Lopes, R.; Rios, F.; Camargo, L.; Anagnostopoulou, A.; Arner, A.; Montezano, A. Vascular smooth muscle contraction in hypertension. Cardiovasc. Res. 2018, 114, 529–539. [Google Scholar] [CrossRef]
  132. Zhu, Y.; Qu, J.; He, L.; Zhang, F.; Zhou, Z.; Yang, S.; Zhou, Y. Calcium in vascular smooth muscle cell elasticity and adhesion: Novel insights into the mechanism of action. Front. Physiol. 2019, 10, 852. [Google Scholar] [CrossRef]
  133. Carlstedt, F.; Lind, L. Hypocalcemic syndromes. Crit. Care Clin. 2001, 17, 139–153. [Google Scholar] [CrossRef]
  134. Klein, G. Phosphate as an adjunct to calcium in promoting coronary vascular calcification in chronic inflammatory states. eLife 2024, 13, e91808. [Google Scholar] [CrossRef]
  135. Edwards, F.; Taheri, A.; Dann, S.; Dye, J. Characterization of cytolytic neutrophil activation in vitro by amorphous hydrated calcium phosphate as a model of biomaterial inflammation. J. Biomed. Mater. Res. Part A 2011, 96, 552–565. [Google Scholar] [CrossRef]
  136. Chen, J.; Sitsel, A.; Benoy, V.; Sepúlveda, M.; Vangheluwe, P. Primary active Ca2+ transport systems in health and disease. Cold Spring Harb. Perspect. Biol. 2019, 12, a035113. [Google Scholar] [CrossRef]
  137. Bers, D.M. Cardiac Excitation-Contraction Coupling. Nature 2002, 415, 198–205. [Google Scholar] [CrossRef]
  138. MacLennan, D.H.; Wong, P.T. Isolation of a High Capacity Ca2+-Binding Protein from Sarcoplasmic Reticulum. Nat. New Biol. 1971, 231, 198–200. [Google Scholar]
  139. Beard, N.A.; Laver, D.R.; Dulhunty, A.F. Calsequestrin and the Calcium Release Channel of Skeletal and Cardiac Muscle. Prog. Biophys. Mol. Biol. 2004, 85, 33–69. [Google Scholar] [CrossRef] [PubMed]
  140. Berridge, M.J. Neuronal Calcium Signaling. Neuron 1998, 21, 13–26. [Google Scholar] [CrossRef]
  141. Chin, D.; Means, A.R. Calmodulin: A Prototypical Calcium Sensor. Trends Cell Biol. 2000, 10, 322–328. [Google Scholar] [CrossRef] [PubMed]
  142. Periasamy, M.; Kalyanasundaram, A. SERCA Pump Isoforms: Their Role in Calcium Transport and Disease. Muscle Nerve 2007, 35, 430–442. [Google Scholar] [CrossRef] [PubMed]
  143. Nilius, B.; Voets, T. The Puzzle of TRPV4 Channelopathies. EMBO Rep. 2013, 14, 152–163. [Google Scholar] [CrossRef]
  144. Blaustein, M.P.; Lederer, W.J. Sodium/Calcium Exchange: Its Physiological Implications. Physiol. Rev. 1999, 79, 763–854. [Google Scholar] [CrossRef]
  145. Brini, M.; Carafoli, E. The Plasma Membrane Ca2+ ATPase and the Plasma Membrane Sodium Calcium Exchanger Cooperate in the Regulation of Cell Calcium. Cold Spring Harb. Perspect. Biol. 2011, 3, a004168. [Google Scholar] [CrossRef]
  146. Pizzo, P.; Lissandron, V.; Capitanio, P.; Pozzan, T. Ca2+ Signalling in the Golgi Apparatus. Cell Calcium 2011, 50, 184–192. [Google Scholar] [CrossRef]
  147. Rizzuto, R.; Pozzan, T. Microdomains of Intracellular Ca2+: Molecular Determinants and Functional Consequences. Physiol. Rev. 2006, 86, 369–408. [Google Scholar] [CrossRef]
  148. Prakriya, M.; Lewis, R.S. Store-Operated Calcium Channels. Physiol. Rev. 2015, 95, 1383–1436. [Google Scholar] [CrossRef] [PubMed]
  149. Pizzo, P.; Pozzan, T. Golgi Ca2+, Messengers and Function. Cell Calcium 2007, 42, 405–412. [Google Scholar] [CrossRef]
  150. Brown, E.M. Extracellular Ca2+ sensing, regulation of parathyroid cell function, and role of Ca2+ and other ions as extracellular (first) messengers. Physiol. Rev. 1991, 71, 371–411. [Google Scholar] [CrossRef] [PubMed]
  151. Hofer, A.M.; Brown, E.M. Extracellular calcium sensing and signalling. In Calcium as a Cellular Regulator; Carafoli, E., Klee, C.B., Eds.; Oxford University Press: Oxford, UK, 2003; pp. 233–261. [Google Scholar]
  152. Conigrave, A.D.; Ward, D.T. Calcium-sensing receptor (CaSR): Pharmacological properties and signaling pathways. Best Pract. Res. Clin. Endocrinol. Metab. 2013, 27, 315–331. [Google Scholar] [CrossRef]
  153. Brennan, S.C.; Thiem, U.; Roth, S.; Aggarwal, A.; Fetahu, I.S.; Tennakoon, S.; Gomes, A.R.; Brandi, M.L.; Bruggeman, F.; Mentaverri, R.; et al. Calcium sensing receptor signalling in physiology and cancer. Biochim. Biophys. Acta Mol. Cell Res. 2013, 1833, 1732–1744. [Google Scholar] [CrossRef]
  154. Hannan, F.M.; Babinsky, V.N.; Thakker, R.V. Disorders of the calcium-sensing receptor and partner proteins: Insights into the molecular basis of calcium homeostasis. J. Mol. Endocrinol. 2018, 61, R1–R22. [Google Scholar] [CrossRef]
  155. Hofer, A.M.; Brown, E.M. Extracellular calcium sensing and signalling. Nat. Rev. Mol. Cell Biol. 2003, 2, 521–527. [Google Scholar] [CrossRef]
  156. Hendy, G.N.; Canaff, L. Calcium-Sensing Receptor Gene: Regulation of Expression. Front Physiol. 2016, 7, 394. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  157. Busic-Pavlek, I.; Dumic-Cule, I.; Kovacevic, L.; Milosevic, M.; Delimar, P.; Korsa, L.; Marusic, Z.; Prutki, M. Calcium-Sensing Receptor Expression in Breast Cancer. Int. J. Mol. Sci. 2023, 24, 11678. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  158. Olszak, I.T.; Poznansky, M.C.; Evans, R.H.; Olson, D.; Kos, C.; Pollak, M.R.; Brown, E.M.; Scadden, D.T. Extracellular calcium elicits a chemokinetic response from monocytes in vitro and in vivo. J. Clin. Investig. 2000, 105, 1299–1305. [Google Scholar] [CrossRef]
  159. Rossol, M.; Pierer, M.; Raulien, N.; Quandt, D.; Meusch, U.; Rothe, K.; Schubert, K.; Schöneberg, T.; Schaefer, M.; Krügel, U.; et al. Extracellular Ca2+ is a danger signal activating the NLRP3 inflammasome through G protein-coupled calcium sensing receptors. Nat. Commun. 2012, 3, 1329. [Google Scholar] [CrossRef] [PubMed]
  160. Lee, G.S.; Subramanian, N.; Kim, A.I.; Aksentijevich, I.; Goldbach-Mansky, R.; Sacks, D.B.; Germain, R.N.; Kastner, D.L. The calcium-sensing receptor regulates the NLRP3 inflammasome through Ca2+ and cAMP. Nature 2012, 492, 123–127. [Google Scholar] [CrossRef] [PubMed]
  161. Canton, J.; Schlam, D.; Breuer, C.; Gütschow, M.; Glogauer, M.; Grinstein, S. Calcium-sensing receptors signal constitutive macropinocytosis and facilitate the uptake of large particles and bacteria by macrophages. Cell. Mol. Life Sci. 2016, 73, 3749–3763. [Google Scholar]
  162. Zhai, T.Y.; Cui, B.H.; Zou, L.; Zeng, J.Y.; Gao, S.; Zhao, Q.; Wang, Y.; Xie, W.L.; Sun, Y.H. Expression and Role of the Calcium-Sensing Receptor in Rat Peripheral Blood Polymorphonuclear Neutrophils. Oxid. Med. Cell. Longev. 2017, 2017, 3869561. [Google Scholar] [CrossRef] [PubMed]
  163. Samakai, E.; Go, C.; Soboloff, J. Defining the Roles of Ca2+ Signals during T Cell Activation. In Signaling Mechanisms Regulating T Cell Diversity and Function; Soboloff, J., Kappes, D.J., Eds.; CRC Press/Taylor & Francis: Boca Raton, FL, USA, 2018; Chapter 10. Available online: https://www.ncbi.nlm.nih.gov/books/NBK532321/ (accessed on 5 December 2024). [CrossRef]
  164. Werner, L.E.; Wagner, U. Calcium-Sensing Receptor-Mediated NLRP3 Inflammasome Activation in Rheumatoid Arthritis and Autoinflammation. Front. Physiol. 2023, 13, 1078569. [Google Scholar] [CrossRef]
  165. Liu, W.; Guo, Y.; Liu, Y.; Sun, J.; Yin, X. Calcium-Sensing Receptor of Immune Cells and Diseases. Cardiovasc. Innov. Appl. 2021, 5, 257–266. [Google Scholar] [CrossRef]
  166. Jäger, E.; Murthy, S.; Schmidt, C.; Hahn, M.; Strobel, S.; Peters, A.; Stäubert, C.; Sungur, P.; Venus, T.; Geisler, M.; et al. Calcium-Sensing Receptor-Mediated NLRP3 Inflammasome Response to Calciprotein Particles Drives Inflammation in Rheumatoid Arthritis. Nat. Commun. 2020, 11, 4243. [Google Scholar] [CrossRef]
  167. Su, Y.; Zhang, Y.; Hu, Z.; He, L.; Wang, W.; Xu, J.; Fan, Z.; Liu, C.; Zhang, H.; Zhao, K. Prokineticin 2 via Calcium-Sensing Receptor Activated NLRP3 Inflammasome Pathway in the Testicular Macrophages of Uropathogenic Escherichia coli-Induced Orchitis. Front. Immunol. 2020, 11, 570872. [Google Scholar] [CrossRef]
  168. Miyazono, K. Transforming Growth Factor-Beta Signaling in Epithelial-Mesenchymal Transition and Progression of Cancer. Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 2009, 85, 314–323. [Google Scholar] [CrossRef]
  169. Wang, L.; Li, J.; Zhong, H.; Qu, Y.; Xi, D.; Tang, N.; He, F. Calcium-Sensitive Receptors-Mediated Macrophage Polarization via the PLC-Ca2+ and cAMP-NF-κB-NLRP3 Inflammasome Signalling Pathways Impact Cardiac Fibroblast Activation through Paracrine Effects. ESS Open Arch. eprints 2024, 65, 06544864. [Google Scholar] [CrossRef]
  170. Liu, J.O. Calmodulin-Dependent Phosphatase, Kinases, and Transcriptional Corepressors Involved in T-Cell Activation. Immunol. Rev. 2009, 228, 184–198. [Google Scholar] [CrossRef] [PubMed]
  171. Scharenberg, A.M.; Humphries, L.A.; Rawlings, D.J. Calcium Signalling and Cell-Fate Choice in B Cells. Nat. Rev. Immunol. 2007, 7, 778–789. [Google Scholar] [CrossRef]
  172. Kadowaki, N.; Antonenko, S.; Lau, J.Y.; Liu, Y.J. Natural Interferon Alpha/Beta-Producing Cells Link Innate and Adaptive Immunity. J. Exp. Med. 2000, 192, 219–226. [Google Scholar] [CrossRef] [PubMed]
  173. Walkon, L.L.; Strubbe-Rivera, J.O.; Bazil, J.N. Calcium Overload and Mitochondrial Metabolism. Biomolecules 2022, 12, 1891. [Google Scholar] [CrossRef] [PubMed]
  174. Clapham, D.E. Calcium signaling. Cell 2007, 131, 1047–1058. [Google Scholar] [CrossRef]
  175. Zaloga, G.P. Hypocalcemia in critically ill patients. Crit. Care Med. 1992, 20, 251–262. [Google Scholar] [CrossRef] [PubMed]
  176. Sheinenzon, A.; Shehadeh, M.; Michelis, R.; Shaoul, E.; Ronen, O. Serum albumin levels and inflammation. Int. J. Biol. Macromol. 2021, 184, 857–862. [Google Scholar] [CrossRef] [PubMed]
  177. Cruz, I.; Barca, J.; Pereira, I.; Meikle, A.; Ruprechter, G. Association between non-esterified fatty acids and calcium concentrations at calving with early lactation clinical diseases, fertility and culling in grazing dairy cows in Uruguay. Prev. Vet. Med. 2024, 230, 106294. [Google Scholar] [CrossRef] [PubMed]
  178. Duran-Güell, M.; Flores-Costa, R.; Casulleras, M.; López-Vicario, C.; Titos, E.; Díaz, A.; Alcaraz-Quiles, J.; Horrillo, R.; Costa, M.; Fernández, J.; et al. Albumin protects the liver from tumor necrosis factor α-induced immunopathology. FASEB J. 2021, 35, e21365. [Google Scholar] [CrossRef]
  179. Levels, J.H.; Marquart, J.A.; Abraham, P.R.; van den Ende, A.E.; Molhuizen, H.O.; van Deventer, S.J.; Meijers, J.C. Lipopolysaccharide is transferred from high-density to low-density lipoproteins by lipopolysaccharide-binding protein and phospholipid transfer protein. Infect. Immun. 2005, 73, 2321–2326. [Google Scholar] [CrossRef]
  180. Cavaillon, J.M.; Adib-Conquy, M.; Fitting, C.; Adrie, C.; Payen, D. Cytokine cascade in sepsis. Scand. J. Infect. Dis. 2003, 35, 535–544. [Google Scholar] [CrossRef]
  181. Morin, E.E.; Guo, L.; Schwendeman, A.; Li, X.A. HDL in sepsis—Risk factor and therapeutic approach. Front. Pharmacol. 2015, 6, 244. [Google Scholar] [CrossRef] [PubMed]
  182. Khovidhunkit, W.; Kim, M.S.; Memon, R.A.; Shigenaga, J.K.; Moser, A.H.; Feingold, K.R.; Grunfeld, C. Effects of infection and inflammation on lipid and lipoprotein metabolism: Mechanisms and consequences to the host. J. Lipid Res. 2004, 45, 1169–1196. [Google Scholar] [CrossRef] [PubMed]
  183. Chandler, T.L.; Westhoff, T.A.; Behling Kelly, E.L.; Sipka, A.S.; Mann, S. Eucalcemia during lipopolysaccharide challenge in postpartum dairy cows: I. Clinical, inflammatory, and metabolic response. J. Dairy Sci. 2023, 106, 3586–3600. [Google Scholar] [CrossRef]
  184. Horst, E.A.; Mayorga, E.J.; Al Qaisi, M.; Abeyta, M.A.; Portner, S.L.; McCarthy, C.S.; Goetz, B.M.; Kvidera, S.K.; Baumgard, L.H. Effects of maintaining eucalcemia following immunoactivation in lactating Holstein dairy cows. J. Dairy Sci. 2020, 103, 7472–7486. [Google Scholar] [CrossRef] [PubMed]
  185. Waldron, M.R.; Nonnecke, B.J.; Nishida, T.; Horst, R.L.; Overton, T.R. Effect of lipopolysaccharide infusion on serum macromineral and vitamin D concentrations in dairy cows. J. Dairy Sci. 2003, 86, 3440–3446. [Google Scholar] [CrossRef]
  186. Seminara, J.A.; Seely, C.R.; McArt, J.A.A. Acute phase responses in clinically healthy multiparous Holsteins with and without calcium dysregulation during the early postpartum period. J. Dairy Sci. 2024, 108, 1930–1939. [Google Scholar] [CrossRef]
  187. Couto Serrenho, R.; Morrison, E.; Bruinjé, T.C.; LeBlanc, S.J. Assessment of systemic inflammation following oral calcium supplementation in healthy postpartum multiparous dairy cows—A randomized controlled trial. JDS Commun. 2023, 5, 134–138. [Google Scholar] [CrossRef]
  188. Aberegg, S.K. Ionized Calcium in the ICU: Should It Be Measured and Corrected? Chest 2016, 149, 846–855. [Google Scholar] [CrossRef] [PubMed]
  189. McArt, J.A.A.; Neves, R.C. Association of transient, persistent, or delayed subclinical hypocalcemia with early lactation disease, removal, and milk yield in Holstein cows. J. Dairy Sci. 2020, 103, 690–701. [Google Scholar] [CrossRef] [PubMed]
  190. Heaney, R.P. Long-latency deficiency disease: Insights from calcium and vitamin D. Am. J. Clin. Nutr. 2003, 78, 912–919. [Google Scholar] [CrossRef] [PubMed]
  191. Garidel, P.; Rappolt, M.; Schromm, A.; Howe, J.; Lohner, K.; Andrä, J.; Koch, M.; Brandenburg, K. Divalent cations affect chain mobility and aggregate structure of lipopolysaccharide from Salmonella minnesota reflected in a decrease of its biological activity. Biochim. Biophys. Acta 2005, 1715, 122–131. [Google Scholar] [CrossRef]
  192. Redeker, C.; Briscoe, W. Interactions between mutant bacterial lipopolysaccharide (LPS Ra) surface layers: Surface vesicles, membrane fusion, and effect of Ca2+ and temperature. Langmuir 2019, 35, 1352–1362. [Google Scholar] [CrossRef] [PubMed]
  193. Ryu, J.K.; Kim, S.J.; Rah, S.H.; Kang, J.I.; Jung, H.E.; Lee, D.; Lee, H.K.; Lee, J.O.; Park, B.S.; Yoon, T.Y.; et al. Reconstruction of LPS transfer cascade reveals structural determinants within LBP, CD14, and TLR4 MD2 for efficient LPS recognition and transfer. Immunity 2017, 46, 38–50. [Google Scholar] [CrossRef]
  194. Drago Serrano, M.E.; de la Garza Amaya, M.; Luna, J.S.; Campos Rodríguez, R. Lactoferrin lipopolysaccharide (LPS) binding as key to antibacterial and antiendotoxic effects. Int. Immunopharmacol. 2012, 12, 1–9. [Google Scholar] [CrossRef]
  195. Wurfel, M.M.; Wright, S.D. Lipopolysaccharide binding protein and soluble CD14 transfer lipopolysaccharide to phospholipid bilayers: Preferential interaction with particular classes of lipid. J. Immunol. 1997, 158, 3925–3934. [Google Scholar] [CrossRef] [PubMed]
  196. Tsukamoto, H.; Takeuchi, S.; Kubota, K.; Kobayashi, Y.; Kozakai, S.; Ukai, I.; Shichiku, A.; Okubo, M.; Numasaki, M.; Kanemitsu, Y.; et al. Lipopolysaccharide (LPS) binding protein stimulates CD14 dependent toll-like receptor 4 internalization and LPS-induced TBK1 IKKϵ IRF3 axis activation. J. Biol. Chem. 2018, 293, 10186–10201. [Google Scholar] [CrossRef]
  197. Uhrig, A.; Banafsche, R.; Kremer, M.; Hegenbarth, S.; Hamann, A.; Neurath, M.; Gerken, G.; Limmer, A.; Knolle, P.A. Development and functional consequences of LPS tolerance in sinusoidal endothelial cells of the liver. J. Leukoc. Biol. 2005, 77, 626–633. [Google Scholar] [CrossRef]
  198. Baranova, I.N.; Vishnyakova, T.G.; Bocharov, A.V.; Leelahavanichkul, A.; Kurlander, R.; Chen, Z.; Souza, A.C.; Yuen, P.S.; Star, R.A.; Csako, G.; et al. Class B scavenger receptor types I and II and CD36 mediate bacterial recognition and proinflammatory signaling induced by Escherichia coli, lipopolysaccharide, and cytosolic chaperonin 60. J. Immunol. 2012, 188, 1371–1380. [Google Scholar] [CrossRef]
  199. Van Bossuyt, H.; Wisse, E. Structural changes produced in Kupffer cells in the rat liver by injection of lipopolysaccharide. Cell Tissue Res. 1988, 251, 205–214. [Google Scholar] [CrossRef]
  200. Tamura, Y.; Osuga, J.; Adachi, H.; Tozawa, R.; Takanezawa, Y.; Ohashi, K.; Yahagi, N.; Sekiya, M.; Okazaki, H.; Tomita, S.; et al. Scavenger receptor expressed by endothelial cells I (SREC I) mediates the uptake of acetylated low-density lipoproteins by macrophages stimulated with lipopolysaccharide. J. Biol. Chem. 2004, 279, 30938–30944. [Google Scholar] [CrossRef] [PubMed]
  201. König, V.; Hopf, U.; Möller, B.; Lobeck, H.; Assmann, G.; Freudenberg, M.; Galanos, C. The significance of high-density lipoproteins (HDL) in the clearance of intravenously administered bacterial lipopolysaccharides (LPS) in mice. Hepatogastroenterology 1988, 35, 111–115. [Google Scholar] [PubMed]
  202. Hersoug, L.G.; Møller, P.; Loft, S. Gut microbiota derived lipopolysaccharide uptake and trafficking to adipose tissue: Implications for inflammation and obesity. Obes. Rev. 2016, 17, 297–312. [Google Scholar] [CrossRef] [PubMed]
  203. Lumeng, C.N.; Bodzin, J.L.; Saltiel, A.R. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J. Clin. Investig. 2007, 117, 175–184. [Google Scholar] [CrossRef]
  204. Kumar, P.; Schroder, E.A.; Rajaram, M.V.S.; Harris, E.N.; Ganesan, L.P. The battle of LPS clearance in host defense vs. inflammatory signaling. Cells 2024, 13, 1590. [Google Scholar] [CrossRef]
Figure 1. The Calci-Inflammatory Network: A complex system of interconnected nodes linking inflammation and calcium regulation in periparturient dairy cows. This figure illustrates the central hypothesis of the Calci-Inflammatory Network as a multidimensional physiological network linking systemic inflammation with calcium homeostasis. The process begins with the entry of bacterial lipopolysaccharide (LPS) into the systemic circulation (1), originating from mucosal sites such as the rumen, mammary gland, uterus, and hoof during postpartum (2), or from prepartum conditions such as subclinical mastitis (3). Once in circulation (4), LPS is recognized by Toll-like receptor 4 (TLR4) on macrophages and neutrophils (5), triggering NF-κB–mediated release of proinflammatory cytokines including IL-1, IL-6, and TNF-α (6). These cytokines interfere with calcium regulation by inhibiting parathyroid hormone (PTH) secretion (7), and consequently kidney calcium reabsorption (8), bone resorption (9), and intestinal absorption (10). Collectively, these processes lead to hypocalcemia, particularly reduced ionized calcium (iCa2+) (11). The drop in iCa2+ is sensed by calcium-sensing receptors (CaSRs) on immune cells (12), which suppresses calcium influx and downstream immune activation, dampening further cytokine release (13) as a negative feedback mechanism. However, if this loop is dysregulated—due to excessive inflammation or delayed calcium mobilization—it results in pathological hypocalcemia and contributes to periparturient diseases such as mastitis, metritis, and laminitis (14) [Figure created using 2025 BioRender in collaboration with Zohaib Saleem]. Note: Downward arrows indicate a decrease, whereas upward arrows indicate an increase.
Figure 1. The Calci-Inflammatory Network: A complex system of interconnected nodes linking inflammation and calcium regulation in periparturient dairy cows. This figure illustrates the central hypothesis of the Calci-Inflammatory Network as a multidimensional physiological network linking systemic inflammation with calcium homeostasis. The process begins with the entry of bacterial lipopolysaccharide (LPS) into the systemic circulation (1), originating from mucosal sites such as the rumen, mammary gland, uterus, and hoof during postpartum (2), or from prepartum conditions such as subclinical mastitis (3). Once in circulation (4), LPS is recognized by Toll-like receptor 4 (TLR4) on macrophages and neutrophils (5), triggering NF-κB–mediated release of proinflammatory cytokines including IL-1, IL-6, and TNF-α (6). These cytokines interfere with calcium regulation by inhibiting parathyroid hormone (PTH) secretion (7), and consequently kidney calcium reabsorption (8), bone resorption (9), and intestinal absorption (10). Collectively, these processes lead to hypocalcemia, particularly reduced ionized calcium (iCa2+) (11). The drop in iCa2+ is sensed by calcium-sensing receptors (CaSRs) on immune cells (12), which suppresses calcium influx and downstream immune activation, dampening further cytokine release (13) as a negative feedback mechanism. However, if this loop is dysregulated—due to excessive inflammation or delayed calcium mobilization—it results in pathological hypocalcemia and contributes to periparturient diseases such as mastitis, metritis, and laminitis (14) [Figure created using 2025 BioRender in collaboration with Zohaib Saleem]. Note: Downward arrows indicate a decrease, whereas upward arrows indicate an increase.
Dairy 06 00022 g001
Figure 2. Hypocalcemia as an AdapiCa2+ modulates the clearance and inflammatory potential of lipopolysaccharide (LPS) originating from mucosal tissues such as the mammary gland, gastrointestinal tract, reproductive tract, and infected hooves (1, 2, 3, 4). LPS traverses the epithelial barrier into the bloodstream (5), where it binds to chelators (e.g., albumin, lactate, and NEFA), which reduce free iCa2+ (7). Under transient or moderate hypocalcemia (10), lower iCa2+ dampens certain calcium-dependent immune pathways, steering LPS toward lipoprotein-mediated clearance via acute-phase high-density lipoproteins containing serum amyloid A (SAA-ap-HDL) (9). Lipopolysaccharidetive Response to Systemic Inflammation. This schematic illustrates how shifts in ionized calcium (-binding protein (LBP) and CD14 (11) facilitate LPS binding to ap-HDL, directing it for detoxification in hepatocytes (possibly contributing to fatty liver) (12) or temporary storage in adipocytes (13). Apolipoprotein A1, dissociated from n-HDL, also binds LPS for renal excretion (14). During mild hypocalcemia, macrophage activation (15) remains subdued, leading to limited release of pro-inflammatory cytokines (IL-1, IL-6, TNF-α). In contrast, normocalcemia (16) supports lamellar LPS aggregation in the presence of iCa2+, enabling macrophages to take up these larger, structurally inert complexes (17). Although macrophages then release cytokines (IL-1, IL-6, TNF-α) (18), the inflammatory response may remain more localized than systemic. Some fraction of LPS is also bound by normal HDL (n-HDL) and cleared by the liver (19). Overall, this figure emphasizes that mild hypocalcemia can serve as an adaptive brake on systemic inflammation by reducing excessive immune-cell activation and directing LPS toward lipoprotein-based clearance, whereas adequate iCa2+ levels favor lamellar LPS aggregation and macrophage uptake. Taken together, these complementary pathways underscore the dynamic role of calcium homeostasis and lipoprotein interactions in balancing LPS clearance under varying inflammatory states. For detailed information on the concentration of LPS (endotoxin) in blood circulation, please refer to Supplementary Table S2. [Figure created using 2025 BioRender in collaboration with Zohaib Saleem (2025) https://BioRender.com/shc8ckv].
Figure 2. Hypocalcemia as an AdapiCa2+ modulates the clearance and inflammatory potential of lipopolysaccharide (LPS) originating from mucosal tissues such as the mammary gland, gastrointestinal tract, reproductive tract, and infected hooves (1, 2, 3, 4). LPS traverses the epithelial barrier into the bloodstream (5), where it binds to chelators (e.g., albumin, lactate, and NEFA), which reduce free iCa2+ (7). Under transient or moderate hypocalcemia (10), lower iCa2+ dampens certain calcium-dependent immune pathways, steering LPS toward lipoprotein-mediated clearance via acute-phase high-density lipoproteins containing serum amyloid A (SAA-ap-HDL) (9). Lipopolysaccharidetive Response to Systemic Inflammation. This schematic illustrates how shifts in ionized calcium (-binding protein (LBP) and CD14 (11) facilitate LPS binding to ap-HDL, directing it for detoxification in hepatocytes (possibly contributing to fatty liver) (12) or temporary storage in adipocytes (13). Apolipoprotein A1, dissociated from n-HDL, also binds LPS for renal excretion (14). During mild hypocalcemia, macrophage activation (15) remains subdued, leading to limited release of pro-inflammatory cytokines (IL-1, IL-6, TNF-α). In contrast, normocalcemia (16) supports lamellar LPS aggregation in the presence of iCa2+, enabling macrophages to take up these larger, structurally inert complexes (17). Although macrophages then release cytokines (IL-1, IL-6, TNF-α) (18), the inflammatory response may remain more localized than systemic. Some fraction of LPS is also bound by normal HDL (n-HDL) and cleared by the liver (19). Overall, this figure emphasizes that mild hypocalcemia can serve as an adaptive brake on systemic inflammation by reducing excessive immune-cell activation and directing LPS toward lipoprotein-based clearance, whereas adequate iCa2+ levels favor lamellar LPS aggregation and macrophage uptake. Taken together, these complementary pathways underscore the dynamic role of calcium homeostasis and lipoprotein interactions in balancing LPS clearance under varying inflammatory states. For detailed information on the concentration of LPS (endotoxin) in blood circulation, please refer to Supplementary Table S2. [Figure created using 2025 BioRender in collaboration with Zohaib Saleem (2025) https://BioRender.com/shc8ckv].
Dairy 06 00022 g002
Figure 3. Tissues and Organs Involved in iCa2+ Buffering and Sequestration During Inflammation. Calcium in the bloodstream exists in three primary forms: protein-bound (~40%), ionized (iCa2+, ~50%), and chelated (~10%). This delicate balance supports essential physiological processes and adapts to systemic inflammation (1). Platelets sequester and release iCa2+ during inflammatory processes to regulate coagulation and immune responses (2). Red blood cells buffer iCa2+ to maintain membrane flexibility and oxygen transport, both of which can be compromised under inflammatory conditions (3). Immune cells depend on iCa2+ buffering for activation, migration, and cytokine release—functions that intensify during inflammation (4). Skeletal muscle cells store and release iCa2+ via the sarcoplasmic reticulum, enabling contractions that may be heightened in inflammatory states (5). Bones act as the largest calcium reservoir, releasing iCa2+ into the bloodstream during inflammation-induced hypocalcemia or storing it in periods of hypercalcemia (6). The kidneys regulate iCa2+ homeostasis through reabsorption or excretion; however, systemic inflammation can impair renal function, leading to abnormal calcium wasting or retention (7). Hepatocytes buffer iCa2+ within the endoplasmic reticulum and mitochondria, playing a critical role in detoxification and metabolic responses to inflammation (8). Neurons rely on iCa2+ buffering for synaptic signaling, while glial cells modulate intra- and extracellular calcium levels during neuroinflammatory states (9). Vascular endothelial cells buffer iCa2+ in response to inflammatory mediators such as histamine and bradykinin, thereby affecting vascular permeability and tone (10). In the extracellular matrix, iCa2+ binds to collagen, elastic fibers, and osteopontin, buffering excess calcium while maintaining tissue integrity under inflammatory stress (11). Finally, smooth muscle cells regulate contraction and relaxation by buffering and releasing iCa2+—processes crucial for preserving vascular integrity and blood flow during inflammation (12) [Figure created using 2025 BioRender in collaboration with Zohaib Saleem; (2025) https://BioRender.com/shc8ckv].
Figure 3. Tissues and Organs Involved in iCa2+ Buffering and Sequestration During Inflammation. Calcium in the bloodstream exists in three primary forms: protein-bound (~40%), ionized (iCa2+, ~50%), and chelated (~10%). This delicate balance supports essential physiological processes and adapts to systemic inflammation (1). Platelets sequester and release iCa2+ during inflammatory processes to regulate coagulation and immune responses (2). Red blood cells buffer iCa2+ to maintain membrane flexibility and oxygen transport, both of which can be compromised under inflammatory conditions (3). Immune cells depend on iCa2+ buffering for activation, migration, and cytokine release—functions that intensify during inflammation (4). Skeletal muscle cells store and release iCa2+ via the sarcoplasmic reticulum, enabling contractions that may be heightened in inflammatory states (5). Bones act as the largest calcium reservoir, releasing iCa2+ into the bloodstream during inflammation-induced hypocalcemia or storing it in periods of hypercalcemia (6). The kidneys regulate iCa2+ homeostasis through reabsorption or excretion; however, systemic inflammation can impair renal function, leading to abnormal calcium wasting or retention (7). Hepatocytes buffer iCa2+ within the endoplasmic reticulum and mitochondria, playing a critical role in detoxification and metabolic responses to inflammation (8). Neurons rely on iCa2+ buffering for synaptic signaling, while glial cells modulate intra- and extracellular calcium levels during neuroinflammatory states (9). Vascular endothelial cells buffer iCa2+ in response to inflammatory mediators such as histamine and bradykinin, thereby affecting vascular permeability and tone (10). In the extracellular matrix, iCa2+ binds to collagen, elastic fibers, and osteopontin, buffering excess calcium while maintaining tissue integrity under inflammatory stress (11). Finally, smooth muscle cells regulate contraction and relaxation by buffering and releasing iCa2+—processes crucial for preserving vascular integrity and blood flow during inflammation (12) [Figure created using 2025 BioRender in collaboration with Zohaib Saleem; (2025) https://BioRender.com/shc8ckv].
Dairy 06 00022 g003
Figure 4. Intracellular Calcium Buffers. Pathways and components involved in intracellular calcium (Ca2+) buffering across various organelles and cellular structures are shown: (1) In muscle cells, the sarcoplasmic reticulum functions as a primary site for Ca2+ sequestration and release, mediated by calcium-binding proteins such as calsequestrin, sarcalumenin, junctate, and triadin; (2) In the cytoplasm, soluble calcium-binding proteins—including calmodulin, calbindin-D28K, calbindin-D9K, and calreticulin—transiently bind free Ca2+, thereby regulating intracellular levels and signaling; (3) Membrane-associated proteins, such as the sarco/endoplasmic reticulum Ca2+-ATPase (SERCA), transient receptor potential (TRP) calcium channels, and the sodium-calcium exchanger (NCX), transport calcium across cellular membranes to maintain overall calcium homeostasis; (4) Calcium exchange between lysosomes and the endoplasmic reticulum occurs via two-pore calcium channels (TPC1/2), supporting both calcium storage and signaling; (5) At the endoplasmic reticulum (ER)-mitochondria junction, calcium transfer is mediated by inositol trisphosphate receptor (IP3R)-VDAC (voltage-dependent anion channel) complexes, with mitochondria buffering calcium through the rapid uptake mode (RaM) to support metabolic functions; (6) At the ER-plasma membrane junction, store-operated calcium entry (SOCE) is controlled by ORAI-STIM1 (Orai-stromal interaction molecule 1) interactions, which replenish ER calcium stores; (7) The Golgi apparatus also serves as a calcium reservoir, contributing to vesicular trafficking and protein modification. Collectively, these systems underscore the intricate and interconnected mechanisms by which calcium is buffered, stored, and mobilized within cells to support vital cellular processes and signaling pathways [Figure created using 2025 BioRender in collaboration with Zohaib Saleem; (2025) https://BioRender.com/shc8ckv].
Figure 4. Intracellular Calcium Buffers. Pathways and components involved in intracellular calcium (Ca2+) buffering across various organelles and cellular structures are shown: (1) In muscle cells, the sarcoplasmic reticulum functions as a primary site for Ca2+ sequestration and release, mediated by calcium-binding proteins such as calsequestrin, sarcalumenin, junctate, and triadin; (2) In the cytoplasm, soluble calcium-binding proteins—including calmodulin, calbindin-D28K, calbindin-D9K, and calreticulin—transiently bind free Ca2+, thereby regulating intracellular levels and signaling; (3) Membrane-associated proteins, such as the sarco/endoplasmic reticulum Ca2+-ATPase (SERCA), transient receptor potential (TRP) calcium channels, and the sodium-calcium exchanger (NCX), transport calcium across cellular membranes to maintain overall calcium homeostasis; (4) Calcium exchange between lysosomes and the endoplasmic reticulum occurs via two-pore calcium channels (TPC1/2), supporting both calcium storage and signaling; (5) At the endoplasmic reticulum (ER)-mitochondria junction, calcium transfer is mediated by inositol trisphosphate receptor (IP3R)-VDAC (voltage-dependent anion channel) complexes, with mitochondria buffering calcium through the rapid uptake mode (RaM) to support metabolic functions; (6) At the ER-plasma membrane junction, store-operated calcium entry (SOCE) is controlled by ORAI-STIM1 (Orai-stromal interaction molecule 1) interactions, which replenish ER calcium stores; (7) The Golgi apparatus also serves as a calcium reservoir, contributing to vesicular trafficking and protein modification. Collectively, these systems underscore the intricate and interconnected mechanisms by which calcium is buffered, stored, and mobilized within cells to support vital cellular processes and signaling pathways [Figure created using 2025 BioRender in collaboration with Zohaib Saleem; (2025) https://BioRender.com/shc8ckv].
Dairy 06 00022 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ametaj, B.N. The Calci-Inflammatory Network: A Paradigm Shift in Understanding Milk Fever. Dairy 2025, 6, 22. https://doi.org/10.3390/dairy6030022

AMA Style

Ametaj BN. The Calci-Inflammatory Network: A Paradigm Shift in Understanding Milk Fever. Dairy. 2025; 6(3):22. https://doi.org/10.3390/dairy6030022

Chicago/Turabian Style

Ametaj, Burim N. 2025. "The Calci-Inflammatory Network: A Paradigm Shift in Understanding Milk Fever" Dairy 6, no. 3: 22. https://doi.org/10.3390/dairy6030022

APA Style

Ametaj, B. N. (2025). The Calci-Inflammatory Network: A Paradigm Shift in Understanding Milk Fever. Dairy, 6(3), 22. https://doi.org/10.3390/dairy6030022

Article Metrics

Back to TopTop