Next Article in Journal
A Consolidated Saccharification, Fermentation, and Transesterification Process (cSFT) Converting Castor Oil to Biodiesel with Cellulose-Derived Ethanol
Previous Article in Journal
The Current Landscape of Modular CAR T Cells
Previous Article in Special Issue
Calmodulin Interaction Interface with Plasma Membrane Ca2+-ATPase Isoforms: An Integrative Bioinformatic Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Intracellular Calcium as a Regulator of Polarization and Target Reprogramming of Macrophages

by
Marina Y. Pogonyalova
,
Daniil Y. Popov
and
Andrey Y. Vinokurov
*
Cell Physiology and Pathology Laboratory, Orel State University, Orel 302026, Russia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(24), 11901; https://doi.org/10.3390/ijms262411901
Submission received: 10 November 2025 / Revised: 6 December 2025 / Accepted: 8 December 2025 / Published: 10 December 2025
(This article belongs to the Special Issue Calcium Homeostasis of Cells in Health and Disease: Third Edition)

Abstract

Macrophage metabolic plasticity providing their polarization towards classically (M1) or alternatively (M2) activated cells is an important element of the initiation, development, and resolving or inflammation-linked pathologies. The prevalence of M1 or M2 types of macrophages during different stages of diseases supports increased inflammation and phagocytosis or tissue repair, respectively. An imbalance leading to a shift toward an M1- or M2-dominant state is associated with a chronic pathological process. This characterizes the regulation of macrophage phenotypes as a prospective strategy in the treatment of various diseases and makes it relevant to a deep understanding of the mechanisms defining cell polarization. According to the central role of calcium signaling in cell metabolism, changes in calcium homeostasis are closely linked to the regulation of polarization. The exact balance between calcium flows across plasma and intracellular membranes provided by a number of receptors and channels, as well as the differences in the calcium-buffering capability of endoplasmic reticulum and mitochondria, are able to influence macrophage polarization towards an M1 or M2 phenotype. This review focuses on the role of the calcium homeostasis system in macrophage functionality and calcium-induced changes in macrophage metabolism that forms the basis of target disease therapy.

1. Introduction

Calcium intracellular signaling system is one of the key players of cell metabolism: physiological calcium concentration changes are linked with the regulation of mitochondrial bioenergetics, lipid metabolism, cytoskeletal modifications, and pathological changes leading to cell death [1,2,3]. The gradients of calcium ions (Ca2+) concentration relative to the plasma and intracellular membranes are the signs of healthy cells. A steady-state cytosolic calcium concentration ([Ca2+]i) (about 100 nM) is 10,000-fold lower compared to the extracellular space, while cell stimulation leads to a 10–100-fold [Ca2+]i increase [4]. The capability of recovery to initial [Ca2+]i, preventing cell damage, is provided by the calcium homeostasis system. It is based on the balance between Ca2+ flows through membranes and the Ca-buffering capability of the cytosol and main intracellular depos–endoplasmic reticulum (ER), mitochondria, and lysosomes. [Ca2+]i increase is provided by extracellular Ca2+ entry though a number of channels, as well as Ca2+ release, mainly from ER [5,6,7,8]. The last is closely linked to cell membrane metabotropic purine receptors P2Y (P2YRs), which are coupled with G-proteins (Dq/G11, Gi/D0, G12/G13 or GS depending on receptor type) [9]. ER calcium release is mainly mediated by the activation of phospholipase C (PLC) by P2Y1R and P2Y6R and the production of inositol triphosphate (IP3), affecting IP3 receptors on the ER membrane [10]. [Ca2+]i decrease is an energy-consuming process of calcium transport into extracellular space through plasma membrane Ca2+-ATPase or into the ER through sarcoplasmic/endoplasmic reticulum Ca2+-ATPase (SERCA) [11,12].
Macrophages are innate immune cells that are present in virtually all tissues, where they perform various functions related to the homeostasis, development, and protection of the body [13,14,15,16]. Tissue-resident macrophages (TRMs) are usually defined as macrophages that colonize tissues during embryonic development and remain there for a long time, often capable of self-renewal in situ [17]. The type of TRM is dependent on the tissue location (Kuppfler cells in liver, Langerhans cells in skin, osteoclasts in bones, microglia in brain, etc.) [18]. In contrast, macrophage-derived macrophages (MDMs) arise from adult bone marrow hematopoietic stem cells via circulating monocytes that can enter tissues, especially during inflammation or the depletion of resident populations [13,19].
During inflammation, resident macrophages accumulate and, in some cases, are replaced by recruited macrophages derived from monocytes. The functional properties of macrophages, both in homeostasis and in disease, are determined not only by their ontogenesis but also by signals from the local tissue microenvironment [20,21,22]. This concept, that origin is independent of activation status, has become a fundamental principle in macrophage biology, emphasizing that two morphologically similar macrophages can have very different histories of origin and functional predispositions. In adulthood, circulating monocytes serve as a source of macrophages that can colonize tissues, especially in response to stimulation. It is important to understand that monocytes themselves are heterogeneous and consist of functionally distinct subgroups [23].
According to the differences in polarizing signals, the role in physiological processes and metabolic specificity macrophages are traditionally divided into two groups—M1 and M2. Classically activated macrophages (M1) are usually induced by microbial products such as lipopolysaccharide (LPS) and interferon-γ (IFN-γ), whereas alternatively activated macrophages (M2) arise in response to interleukin-4 (IL-4) and interleukin-13 (IL-13)—cytokines produced by anti-inflammatory cells (Th2-lymphocytes, innate lymphoid cells type 2) [24]. In addition, one more macrophage group includes the main tumor-infiltrating immune cells, named tumor-associated macrophages (TAMs) [25].
M1 macrophages are characterized by a high production of pro-inflammatory cytokines, including tumor necrosis factor-α (TNF-α), interleukin-1b (IL-1b), and interleukin-6 (IL-6), as well as chemokines such as CXCL9 and CXCL10 [26,27,28]. A key biochemical feature is the activation of induced nitric oxide synthase (iNOS), which leads to the formation of nitric oxide (NO) and reactive oxygen species (ROS), which provide antimicrobial and antitumor activity [29]. From the point of view of metabolism, M1 polarization is associated with increased glycolysis and impaired activity of the tricarboxylic acid (TCA) cycle. The accumulation of metabolites such as succinate stabilizes hypoxia-induced factor-1a, further enhancing the expression of pro-inflammatory genes [30]. At the same time, fatty acid oxidation and the oxidative phosphorylation (OXPHOS) of mitochondria are suppressed, which leads to metabolic reprogramming with functional specialization [31]. Functionally, M1 macrophages play a central role in protecting the body from bacteria, viruses, and protozoa, exerting a bactericidal effect and coordinating the involvement of other immune cells. In the context of pathology, they contribute to acute inflammation and tissue damage when their activity is prolonged [32].
Extensive studies identified variability in alternatively polarized macrophages that allow us to divide them into four main groups—M2a, M2b, M2c, and M2d [33,34,35]. While the first three groups are associated with tissue repair and healing with anti-inflammatory release, the fourth group is responsible for scar formation and fibrosis releasing as anti- and pro-inflammatory cytokines [33]. M2 macrophages are characterized by the expression of common receptors such as CD206 (C-Type Mannose Receptor 1) and CD163 (Hemoglobin–Haptoglobin Scavenger Receptor), as well as enzymatic markers such as arginase-1 [35]. In contrast to the pro-inflammatory functions of M1 cells, M2 macrophages contribute to the elimination of inflammation and the restoration of tissue homeostasis, and also play a central role in angiogenesis and wound healing [36,37].
The metabolic profile of M2 macrophages differs significantly from that of M1 cells. Instead of aerobic glycolysis, M2 polarization is associated with the increased activity of TCA and OXPHOS, and enhanced fatty acid oxidation [38]. This metabolic orientation provides the body with energy and biosynthetic precursors necessary to maintain tissue repair functions.

2. Macrophage Polarization in Progression and Resolution of the Pathologies

Macrophage polarization is an evolutionarily proven instrument in inflammation-linked disease progression and resolution [39,40,41]. While M1 cells are responsible for initiating and sustaining inflammation through releasing pro-inflammatory cytokines and the production of ROS, M2 cells have an anti-inflammation profile including wound healing and tissue repair [40]. So, disease resolution is a result of a well-orchestrated dynamic balance between macrophage types on different stages of the pathology, which is determined by the processes of cell polarization and reprogramming [42,43,44]. Imbalance in macrophages ratio and a persistent shift toward an M1- or M2-dominant state contributes to the pathogenesis of multiple diseases.
The prevalence of M1 macrophages is a sign of chronic inflammation revealed in several pathologies. In atherosclerosis, an increase in pro-inflammatory cells is associated with the growth and instability of plaques and foam cell necrosis with disturbances in the apoptosis-based mechanism of defective cell removal [45]. A widely used model of atherosclerosis (Apolipoprotein E double knockout mice, ApoE-KO) showed that early atherosclerotic plaques were enriched with M2-like macrophages, which promoted the proliferation of smooth muscle cells in vitro, whereas, in elderly mice, a transition from an M2 to M1 phenotype was observed, which is due to the repolarization of macrophages in the lesion. A correlation of lesion progression with the dominance of the M1 phenotype was also determined [46]. High levels of pro-inflammatory cytokine secretion by M1 is a part of the acute or chronic development of autoimmune diseases, including rheumatoid arthritis, systemic lupus erythematosus, Behcet’s disease, multiple sclerosis, lupus nephritis, and multiple sclerosis [34,47,48]. An increased M1 and decreased M2 polarization macrophage level is detected in obesity and type 2 diabetes, indicating that they are chronic inflammatory pathologies [49,50]. Interestingly, this imbalance, coupled with glycemia, leads to the severe development of some other pathologies, including infections [51]. Yao et al., using a Parkinson’s disease (PD) model, C57BL/6 mice treated with methyl-4-phenyl-1,2,3,6-tetrahydropyridine, showed that the microglial activation of the M1 phenotype increases and M2 is suppressed during aging, which correlates with more severe neurodegeneration [52].
M2 macrophage-associated pathology progression is mostly linked with the ability of this type of macrophage to increase cell proliferation, stimulating fibrosis processes and scar formation [53,54,55,56,57]. The infiltration of tumors with M2 macrophages, as well as TAMs with an M2-like phenotype, is responsible for the increased proliferation, migration ability, and epithelial–mesenchymal transition of cancer cells [53,54,55,58,59,60], making macrophages a target in complex cancer therapy. Fibrosis stimulated by M2-type cells leads to pathology progression in the narrowing of the urethral lumen [61], cardiac tissue modeling after myocardial infarction [62], pulmonary fibrosis [63,64,65], and systemic sclerosis [66].
A significant role of M1/M2 macrophage balance in health and disease makes cell polarization and reprogramming a prospective way for the therapy of different pathologies. A deep understanding of the processes regulating monocyte-to-macrophage transformation is needed for this aim. In our opinion, the homeostasis of Ca2+ which mediates and links many intracellular processes can be a target in macrophage phenotype regulation.

3. System of Calcium Homeostasis in Macrophages

Calcium signaling plays a key role in the immune response during infections and inflammation. Calcium promotes cell survival by activating transcription factors, cell proliferation, and the development of phagolysosome fusion [6,67]. Macrophage [Ca2+]i regulation is provided by a well-orchestrated system of receptors and channels of plasma membrane and cell organelles (Figure 1).
Ca2+ influx from extracellular space is mediated by a number of channels opening to external and internal stimuli. External signal-stimulated channels are mostly presented by the ATP-dependent purinergic ionotropic receptors P2X4R and P2X7R and some members of the transient receptor potential (TRP) channel super-family (TRPC1, TRPV2, TRPM2) [68,69,70,71,72,73,74]. Despite their non-excitable character, macrophages also express some voltage-dependent channels (i.e., L-type calcium channels) [75,76].
Another way of [Ca2+]i increase is linked with ions released from intracellular depos, mostly from ER and lysosomes. As in other cell types, Ca2+ accumulation in the ER is mediated by SERCA, while ryanodine (Ry-) and inositol-3-phosphate (IP3-) receptors are responsible for Ca2+ release [77,78]. One of the most studied ways of ER calcium release is associated with the activation of P2YRs [68], mostly P2Y2R and P2Y6R coupled to Gq protein (although there is evidence of the presence of other types of receptors in macrophages, in particular P2Y14R) [79,80,81,82]. ER Ca2+ release is also linked with a lesser known inducer, i.e., bitter taste receptors (T2R), leading to a low-level calcium signals through IP3R activation [83]. ER calcium efflux explains [Ca2+]i’s experimentally registered oscillations, which can mostly be prevented by using IP3R or RyR antagonists or by the removal of extracellular Ca2+ [84,85,86,87,88,89,90].
ER Ca2+ reflux is closely linked with an important immune cell signaling pathway—Ca2+ entry through the plasma membrane via calcium release-activated channels (CRAC) [91], which is called store-operated calcium entry (SOCE) [92,93,94,95]. CRACs are composed of Orai proteins, which form the channel pore and are activated by stromal interaction molecules STIM1 and STIM2. Located in the ER membrane, STIM1 and STIM2 perform a dual function: sensing Ca2+ concentration in the ER and activating CRACs for ER refilling [96,97].
Despite their relatively small size, lysosomes play an important role in calcium signaling due to a wide cell distribution and, therefore, provide the possibility for local [Ca2+] regulation. To date, the list of channels responsible for Ca2+ efflux from lysosomes includes the mucolipin subfamily of transient receptor potential (in macrophages, mostly TRPML1), two-pore channels (TPC2), and ATP-gated P2X4R [98,99]. However, the mechanism of lysosome Ca2+ refilling is a debatable question. Due to their high acidity, Ca2+/H+ exchangers, i.e., LCI encoded by TMEM165, may drive pH-dependent Ca2+ uptake into lysosomes [98,100]. Another known way is closely linked with SOCE and ER-to-lysosomes Ca2+ transport [101].
While mitochondrial Ca2+ uptake is primarily mediated by mitochondrial calcium uniporter (MCU), which is regulated by two subunits—MICU1 and MICU2 [84]—Ca2+ release is linked with the Na+/Ca2+ exchanger (NCLX) and leucine zipper–EF hand-containing transmembrane protein 1 (LETM1) [102,103]. One more mechanism of mitochondrial Ca2+ release is linked with the short-term opening of the mitochondrial permeability transition pore (mPTP) [104,105].
No less important for mitochondrial calcium buffering is a close connection of mitochondria with ER through mitochondria-associated membranes (MAMs), forming by RyR and IP3R and mitochondrial voltage-dependent anion channels (VDAC) [106,107,108]. The close contact of ER and outer mitochondrial membrane forming MAM is provided by tethering proteins including voltage-dependent anion channels (VDAC), Mitofusin-1/2, protein tyrosine phosphatase-interacting protein-51 (PTPIP51), the Fission 1 of mitochondria and IP3R, Mitofusin-2, vesicle associated membrane protein-associated protein B (VAPB), and B cell-associated protein 31 (Bap31) of ER [109]. Among the number of functions [110,111,112], Ca2+ transport from the ER to the mitochondrial matrix mediated by IP3R, VDAC, and chaperone protein glucose-regulated protein 75 (GRP75) [113] play a pivotal role in mitochondria function regulation: a moderate load of mitochondrial matrix with Ca2+ stimulates some of the enzymes (pyruvate dehydrogenase, α-ketoglutarate dehydrogenase, isocitrate dehydrogenase), leading to metabolism enhancement, an increase in mitochondrial membrane potential, and ATP production [114]. However, extreme mitochondrial Ca2+ concentration increase leads to enhanced ROS production, the collapse of the electron transport chain function which is a trigger of prolonged mPTP opening and cell apoptosis [115,116].

4. Calcium Signaling and Macrophage Functions

The calcium homeostasis system determinates macrophage functionality. In monocytes and macrophages, P2Y2Rs play an important chemotactic role in the detection of apoptotic cells releasing ATP [117]. A study on the human monocyte cell line THP-1 showed that treatment of the cells with LPS induced rapid ATP release from the cells, which was blocked by the knockdown of SLC17A9 (vesicular nucleotide transporter, VNUT), which is responsible for ATP exocytosis [82,118]. ATP release via exocytosis also activates P2Y11R, leading to macrophage activation and cytokine release [82]. At the same time, the elevation of [Ca2+]i coupled with the activation of G proteins is responsible for the fusion of docked macrophage vesicles and the regulation of macrophage secretion [119]. Furthermore, a role for the G protein-coupled calcium-sensing receptor (CaSR), associated with cardiovascular diseases, has been shown [120]. Elevated extracellular calcium levels can activate CaSR, initiate the corresponding ion channels, and promote calcium entry into the cell. This, in turn, leads to ER calcium release, stimulates inflammasome assembly, activates the effector protein caspase-1, and, after maturation, the release of the proinflammatory cytokine IL-1β [121]. STIM1 has been reported to be essential for FcγR-mediated calcium entry, phagocytosis, inflammatory cytokine production, and autoimmune inflammation [122], which shows a possible role of CaSR in macrophage SOCE. Disruption in lysosomal calcium regulation was linked with IL-1β release, indicating lysosomes as a player in the priming and assembly phase of NLRP3 inflammasome development [123]. Cell injury mediated by extracellular Ca2+ influx was shown in silica-exposed alveolar macrophages [124]. The same toxicity model with [Ca2+]i increase led to irreversible injury of mouse macrophage-like cell line P388D1, regardless of mitochondrial uncoupling or ATP content decrease [125].
Although calcium-dependence of different membrane fusion processes (i.e., release of neurotransmitters in brain cells or insulin in pancreatic beta cells) [126,127] is well-known, the mechanism of the link between Ca-signals and phagocytosis in macrophages has been discussed for a long time [128]. It was shown that, unlike neutrophils, phagosome–lysosome fusion in macrophages is Ca-independent and develops even in very low calcium concentrations [129]. The same results were observed in a model of IgG-coated erythrocytes [130], even in the case of M1-polarized macrophages [131]. Also, no change in the internalization of non-functionalized polystyrene beads in different extracellular calcium concentrations, as well after SERCA inhibitor thapsigargin action, was detected by E. Diler et al. [132]. Nevertheless, the activation and development of phagocytosis is accompanied by calcium signalization, showing its regulation role in the processes after phagosome formation. [Ca2+]i increase was detected as a result of FcγR receptor activation and linked with ions released from the ER [130,131,133]. In addition, spontaneous and transient changes in calcium activity in Langerhans cells is an important mechanism of skin renewal through the engulfment and phagocytosis of damaged cells, like keratinocytes [134].
Phagosomal calcium flux associated with macrophage inositol-requiring enzyme 1α activation enhances fungicidal activity by promoting phagosome maturation [135]. The efficiency of phagocytosis was also linked with ATP-based calcium signaling mediated by P2X4R and P2X7R [136]. It is noteworthy that the inhibition of calcium signaling leading to the alteration of phagosome–lysosome fusion promoted intracellular mycobacterial tuberculosis survival that was inversed by ionomycin application [137]. Calcium signals, both from extra- and intracellular sources, was required for the recruitment of Golgi-derived vesicles to the sites of different cargo uptakes and effective phagocytosis [138]. Ca-release during the fusion of lysosomes and plasma membrane is a part of the mechanism of large extracellular particle phagocytosis mediated by the activation of the big conductance Ca2+-activated potassium channel [139]. The importance of Ca2+ for NLRP3 inflammasome activation also defines the great role of intracellular calcium depos in phagocytosis. For example, MCU-mediated Ca2+ uptake attenuates phagosomal membrane repair and promotes NLRP3 inflammatory response [140]. As was shown by S. Tedesco et al., MCU knockdown led to phagocytosis inhibition associated with the reduced M2-polarizaton [141]. The close relationship between NLRP3 inflammasome activation and ER calcium release associated with ATP stimulation and mediated by C/EPB homologous protein was shown by T. Murakami et al. [142]. Ca2+ release from the intracellular store linked with T2R activation led to NO production and enhanced phagocytosis through cGMP and cAMP forming [83].

5. Calcium Homeostasis: M1/M2 Macrophage Specificity

Considering the role of Ca2+ in macrophage polarization, it is important to distinguish the difference in basal (steady-state) concentration and stimulation-based change which determine M1 or M2 macrophage calcium specificity (Figure 2).
LPS and IFN-γ leading to M1-like macrophage polarization were responsible for chronic basal [Ca2+]i increase, leading to a high production of IL-6, MCP-1, TNF, IFN-γ, IL-10, and IL-1β [95]. The same results were obtained for TRMs (microglia), where chronic basal [Ca2+]i elevation is a factor of LPS-stimulated NO production and the release of certain chemokines and cytokines indicates pro-inflammatory conditions [143]. Increased inflammation and decreased bacterial colonization during Helicobacter pylori was also revealed in a Trpm2−/− mice model exhibiting macrophage calcium overload and enhanced NADPH oxidase activity [70]. One of the reasons for the increased [Ca2+]i influence on macrophage polarization is linked with subsequent mitochondrial disfunction linked with calcium overload and activating the pro-inflammatory phenotype with enhanced macrophage glycolysis [144], elevated ROS production, and impairment of the ability of mitochondria to buffer intracellular calcium oscillations [145].
According to experimental data, an increase in basal [Ca2+]i leading to preferential M1 polarization may be a sequence of different processes. One of them is SOCE, whose inhibition significantly decreases the release of pro-inflammatory cytokines [95]. Another source of elevated [Ca2+]i is ER calcium release. Its inhibiting also caused an impairment in macrophage recruitment and pro-inflammatory phenotype in a model of Zebrafish wound healing [146]. However, it is important to note that ER stress associated with decreased Ca2+ influx through the Orai-TRPC1-STIM1 complex was linked with a promoted M1 phenotype and increased phagocytosis [147]. One more cause of elevated cytosolic [Ca2+] leading to inflammaging and linked with age is a time-dependent decrease in MCU and MICU1 expression decreasing mitochondrial calcium uptake [84]. TRPC1 deficiency prevents macrophages from producing IFN-γ- or Klebsiella pneumonia infection-induced M1 inflammatory mediators [148]. This means that TRPC1 expression influences macrophage plasticity, and TRPC1-mediated calcium influx is necessary for macrophage polarization toward the M1 inflammatory phenotype [148]. This conclusion is confirmed by other authors [149], who have found that, while cells with an M1 phenotype are mostly dependent on TRPC1 Ca-entry in naïve or M2 macrophages, this process is preferentially associated with Orai channels.
The level of stimulated calcium signal is no less important for macrophage polarization. Purinergic receptors are of interest in this aspect. Comparing P2R profiles, J. Merz et al. have concluded that the principle difference between P2-based calcium signaling in M1 and M2 macrophages is the increased [Ca2+]i change in anti-inflammatory cells after ADP application, which is mostly linked with P2Y1R and P2Y6R receptors [81]. However, the polarizing role of P2XRs is also known. While P2X4Ractivation led to the transition from acute to persistent inflammation through the phosphorylation of p38MAPK, physical exercise-based PPARγ increased activity was associated with the reverse effect and inflammation resolve [150]. P2X7R, which is highly expressed in immune cells, plays a significant but contradictory role in macrophage plasticity. On the one hand, a deficiency in P2X7R, which is highly expressed in TAMs, decreases M2-like macrophage polarization, showing the way for lung cancer therapy [71]. In addition, the increased expression of this receptor was linked with prevalent M2 macrophage polarization in a study of macrophages’ role in chronic heart allograph rejection [151]. On the other hand, during ATP-influenced spontaneous M2 polarization, P2X7Rlevel downregulated [152]. Although the activation of P2X7R induces the assembly of NLRP3 inflammasome and the release of pro-inflammatory cytokines, it is also linked with inflammation resolution through the release of anti-inflammatory proteins [153]. It is interesting that P2X7R plays an important role in macrophage M1-to-M2 reprogramming [73]. While in M1 cells the action of ATP on the receptor leads to the release of pro-inflammatory IL-1β and NLRP3 inflammasome activation, in intermediate states (during M1-to-M2 transition) the same ATP influence uncouples NLRP3 inflammasome activation and inhibits IL-1β release. It should be noted that the last is not inherent to M2 macrophages’ state. The dual role of P2X7Rin inflammation was also shown by E. Townsend et al. in a study of alveolar macrophages obtained from patients with asthma. It was found that P2X7R signaling is linked with the release of pro-inflammatory or pro-resolving lipid mediators [154].
The M1 macrophage polarization route is closely linked to intracellular [Ca2+]i oscillations. The LPS stimulation effect was significantly decreased after the inhibition of FcRγ induced calcium oscillations in bone marrow macrophages [85]. P. Hanley et al. have shown that, stimulated by moderate ATP concentration (10 μM), calcium oscillations mediated by P2Y2R and PLC activation lead to an IL-6 transcription increase [86]. In a study [78], the knockout of TRIM family protein MG53 was linked with RyR-linked Ca2+ oscillations and subsequent interferon-β release. The effect of bioceramics for treating bones’ effect on the subsequent rise and decline in [Ca2+]i and calcium oscillations was shown by J. Zhao [87]. The first stage of bioceramics degradation led to an increase in [Ca2+]i and the stimulation of M1 macrophage polarization, but a decrease in extracellular calcium promotes M1-to-M2 transition.
The decreased Ca2+ influx caused by the blockade of the K+ Kir2.1 channel or membrane depolarization was responsible for the suppression of M1-like but increased M2-like macrophage polarization through the CaMK II/ERK/NF-κB pathway, indicating Kir2.1 as a potential target in modulating inflammatory processes [155].
The dependence of macrophage polarization on extracellular Ca-signaling can be illustrated by the study of the cells of patients with rheumatoid arthritis [156], where calcium-macrophages with excessive, constitutive SPP1/osteopontin production and a strong pro-inflammatory cytokine response due to CaSR activation were identified.
The role of mPTP, which is calcium-dependent in macrophage phenotype regulation, is of interest. The inhibition of mPTP formation by cyclosporine A significantly decreases cytoplasmic mitochondrial DNA (mtDNA) release and the inflammatory response in macrophages deficient in mitochondrial membrane protein prohibitin 1 [157]. The inhibition of cyclophilin D, one of the mPTP components, also alleviated links with thyroid-stimulating hormone receptor pro-inflammatory polarization and oxidative stress in primary peritoneal macrophages and RAW264.7 cells [158]. The positive correlation of the LPS-based macrophage M1 polarization route with mPTP increase was shown in vitro in a culture of alveolar macrophages [159].
The data of the macrophage type specificity of MAMs are rather contradictory. On the one hand, it is shown that MAMs play an important role in inflammaging: while inactivated NRLP3-inflammasome is linked with the ER but after activation it migrates to the contact site of the ER and mitochondria [108,160]. In addition, a reduced MAMs formation is linked with improved OXPHOS and the normalization of mitochondrial Ca2+ homeostasis was shown in the model of the deficiency of connexin 43, which promotes M1 macrophage polarization [161]. On the other hand, according to the results of IP3R–VDAC interaction measurements, LPS+IFN-γ stimulated macrophages have a significantly reduced level of MAMs [162]. Due to the responsibility of IP3R–VDAC interaction for ER-to-mitochondria Ca2+ transport, there is a decrease in the above-mentioned stimulation of mitochondrial bioenergetics that is a feature of pro-inflammatory macrophages.

6. Calcium Homeostasis Regulation in Macrophage Reprogramming: Perspectives for Therapy

Macrophage metabolic plasticity makes it possible to use different approaches for cell polarization regulation, which seems to be a prospective way for the resolving of macrophage-linked pathologies of different origins. For example, Guiducci et al. reported that the cell culture of MCA38 (H-2b), colon carcinoma obtained from primary cultures, switches form of infiltrating macrophages from M2- to M1-like (high levels of IL-12/iNOS), causing hemorrhagic necrosis and tumor destruction [163]. Hagemann et al. report that NF-κB (IKKβ) inhibition, specifically in TAMs, reprograms them to an M1 cytotoxic profile (IL-12high/Arg1low/NOS2high), restores antitumor activity, and regresses advanced ovarian tumors in vivo [164]. Sanson et al. showed the possibility of a reverse transition from M1 to M2 in mouse atherosclerotic plaque when exposed to HDL through STAT6 phosphorylation, thereby causing plaque regression [165]. In a PD model, rats with 6-OHDA damage treated with glucocorticoids targeting CD163 (a marker of M2 macrophages) shifted brain myeloid cells towards anti-inflammatory (M2-like) phenotypes and protected dopaminergic neurons of the substantia nigra [166].
According to the above analysis of the literature, calcium homeostasis regulation is one of the instruments for the polarization route regulation or remodeling of cell phenotypes as a strategy for the release of pathologies linked with macrophage type imbalance.
Some of the known studies are based on the regulation of extracellular Ca2+ concentration, possibly influencing CaSR and CaSR-linked intracellular calcium signalization. An alginate-based calcium release system reinforcing the M1-macrophage phenotype and increasing pro-inflammatory cytokines was studied by Y. Abib et al. [167]. It is interesting that alginate-only was responsible for the formation of pro-repair ability and the increase in the phagocytic activity of cells. The positive correlation of cell microenvironment ions (including Ca2+) concentration with a shift toward M1-like macrophage phenotypes was shown in a study of biocompatible materials [168,169]. The sequential regulation of macrophage phenotypes by the release of Ca2+ and Zn2+ from the hydrogel of acrylate-modified engineered protein with oxidized sodium alginate was used for the promotion of M1 polarization for infection inhibition, and then M2 polarization for the promotion of osteogenic differentiation during the restoration of infected bone defects [170].
The other findings are directly linked with intracellular [Ca2+]i regulation. Q. Zhiao et al. suggested lysosomal pH-sensitive ZIF-8 nanoparticles releasing ions (Ca2+ and Zn2+) from a metal–organic matrix into lysosomes, leading to the restoration of impaired phagocytosis [171]. The usage of calcium-chelating mesoporous silica nanoparticles demonstrated the reverse of the pro-inflammatory phenotype of mouse bone marrow-derived macrophages due to [Ca2+]i regulation and the prevention of mitochondrial Ca2+ overload that can be used for the osteoarthritis therapy [145]. Near-infrared-regulating nanoparticles with the ability to increase or decrease [Ca2+]i were shown for predominant macrophage polarization into M1- or M2-type, respectively [172]. The possibility of macrophage function improvement in chronic obstructive pulmonary disease by a change in calcium homeostasis was shown in the model of MDMs incubated with Haemophilus influenzae [173]. An increase in external Ca2+ concentration improved the phagocytosis, cytokine secretion, and cell surface expression of bacterial recognition receptors. The activation of transient receptor potential vanilloid type 4 (TRPV4), leading to a [Ca2+]i increase, triggered the activation of the transcription factor CREB and an increased production of IL-10, associated with the anti-inflammatory macrophages phenotype [174]. The inhibition of osteoclastogenesis in rheumatoid arthritis therapy by cytotoxic T-lymphocyte antigen 4-immunoglobulin was linked with decreased [Ca2+]i oscillations and LPS-induced bone resorption [85].
Since mitochondria are one of the key regulators of cell metabolism and, therefore, macrophage plasticity [145,175,176], and also play an important role in calcium homeostasis regulation, these organelles are the focus of macrophage-regulation studies. While some of the studies are directed towards complex macrophage metabolism change, the others are linked with mitochondrial calcium buffering properties. The inhibition or knockdown of MCU preventing mitochondria calcium uptake may be used for significantly reducing M2-like polarization without changing the level of M1-macrophages that can be used in the therapy of pathologies with stimulated cell proliferation or fibrosis [116]. It was demonstrated that inhibiting ER–mitochondria coupling with reducing mitochondrial calcium overload and shifting macrophage metabolism from glycolysis to OXPHOS can be achieved by amorphous calcium zinc phosphate nanoparticles [177]. The dominant-negative mitochondrial calcium uniporter subunit regulation is capable of M1-to-M2 reprogramming in macrophages, infiltrating damaged skeletal muscle, which can be useful in muscle regeneration therapy [178]. According to the significant role of mPTP in mitochondrial metabolism and the release of pro-inflammatory and pro-apoptotic factors in cytosol, its regulation is an expected target in macrophage phenotype regulation for therapy [157,179,180]. The anti-inflammatory effect linked with mPTP inhibition was shown in a study of tanshinone IIA, an active ingredient of Salvia miltiorrhiza [159].

7. Conclusions

The system of intracellular calcium homeostasis plays an important role in determining macrophage phenotype and the mechanisms of cell reprogramming. The close relationship of extracellular Ca2+ with intracellular calcium depos mediated by numerous receptors and transport systems provides macrophage functionality and the role of switching between M1- and M2-states in the development and resolving of pathologies. Disturbance in this well-balanced system leads to a pathological prevalence of a definite cell type with the development of a chronic inflammation process or undesired cell proliferation and fibrosis. Different macrophage types are characterized by specific calcium-linked processes and regulators that make it possible to shift cell type in a desired direction by the regulation of steady-state calcium concentration or the character of its stimulated change.

Author Contributions

Conceptualization, A.Y.V.; writing—original draft preparation, A.Y.V., M.Y.P., D.Y.P.; writing—review and editing, A.Y.V., M.Y.P., D.Y.P.; supervision, A.Y.V.; project administration, A.Y.V. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Russian Science Foundation, grant number 22-15-00317-П (https://rscf.ru/en/project/22-15-00317/, accessed on 5 December 2025).

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
CaSRCalcium-sensing receptor
CRACCalcium release-activated channel
EREndoplasmic reticulum
GPCRG protein-coupled receptor
IP3RInositol-3-phosphate receptor
LPSLipopolysaccharide
MAMsMitochondria-associated membranes
MCUMitochondrial calcium uniporter
MDMs Macrophage-derived macrophages
NCLXNa+/Ca2+ exchanger
OXPHOSOxidative phosphorylation
PDParkinson’s disease
RyRRyanodine receptor
SOCEStore-operated calcium entry
TAMsTumor-associated macrophages
TCATricarboxylic acid cycle
TRMsTissue-resident macrophages
VDACvoltage-dependent anion channel
VNUTvesicular nucleotide transporter

References

  1. Wang, Y.; Tao, A.; Vaeth, M.; Feske, S. Calcium Regulation of T Cell Metabolism. Curr. Opin. Physiol. 2020, 17, 207–223. [Google Scholar] [CrossRef]
  2. Rimessi, A.; Pedriali, G.; Vezzani, B.; Tarocco, A.; Marchi, S.; Wieckowski, M.R.; Giorgi, C.; Pinton, P. Interorganellar Calcium Signaling in the Regulation of Cell Metabolism: A Cancer Perspective. Semin. Cell Dev. Biol. 2020, 98, 167–180. [Google Scholar] [CrossRef] [PubMed]
  3. Hu, T.; Liu, C.-H.; Lei, M.; Zeng, Q.; Li, L.; Tang, H.; Zhang, N. Metabolic Regulation of the Immune System in Health and Diseases: Mechanisms and Interventions. Signal Transduct. Target. Ther. 2024, 9, 268. [Google Scholar] [CrossRef] [PubMed]
  4. Bagur, R.; Hajnóczky, G. Intracellular Ca(2+) Sensing: Its Role in Calcium Homeostasis and Signaling. Mol. Cell 2017, 66, 780–788. [Google Scholar] [CrossRef]
  5. Daverkausen-Fischer, L.; Pröls, F. Regulation of Calcium Homeostasis and Flux between the Endoplasmic Reticulum and the Cytosol. J. Biol. Chem. 2022, 298, 102061. [Google Scholar] [CrossRef] [PubMed]
  6. 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]
  7. Brini, M.; Calì, T.; Ottolini, D.; Carafoli, E. Intracellular Calcium Homeostasis and Signaling. Met. Ions Life Sci. 2013, 12, 119–168. [Google Scholar] [CrossRef]
  8. Sukhorukov, V.N.; Khotina, V.A.; Bagheri Ekta, M.; Ivanova, E.A.; Sobenin, I.A.; Orekhov, A.N. Endoplasmic Reticulum Stress in Macrophages: The Vicious Circle of Lipid Accumulation and Pro-Inflammatory Response. Biomedicines 2020, 8, 210. [Google Scholar] [CrossRef]
  9. Rueschpler, L.; Schloer, S. Beyond the Surface: P2Y Receptor Downstream Pathways, TLR Crosstalk and Therapeutic Implications for Infection and Autoimmunity. Pharmacol. Res. 2025, 219, 107884. [Google Scholar] [CrossRef]
  10. Cekic, C.; Linden, J. Purinergic Regulation of the Immune System. Nat. Rev. Immunol. 2016, 16, 177–192. [Google Scholar] [CrossRef]
  11. Krebs, J. Structure, Function and Regulation of the Plasma Membrane Calcium Pump in Health and Disease. Int. J. Mol. Sci. 2022, 23, 1027. [Google Scholar] [CrossRef]
  12. Viskupicova, J.; Espinoza-Fonseca, L.M. Allosteric Modulation of SERCA Pumps in Health and Disease: Structural Dynamics, Posttranslational Modifications, and Therapeutic Potential. J. Mol. Biol. 2025, 437, 169200. [Google Scholar] [CrossRef] [PubMed]
  13. Hashimoto, D.; Chow, A.; Noizat, C.; Teo, P.; Beasley, M.B.; Leboeuf, M.; Becker, C.D.; See, P.; Price, J.; Lucas, D.; et al. Tissue-Resident Macrophages Self-Maintain Locally throughout Adult Life with Minimal Contribution from Circulating Monocytes. Immunity 2013, 38, 792–804. [Google Scholar] [CrossRef] [PubMed]
  14. Epelman, S.; Lavine, K.J.; Randolph, G.J. Origin and Functions of Tissue Macrophages. Immunity 2014, 41, 21–35. [Google Scholar] [CrossRef] [PubMed]
  15. Bajgar, A.; Krejčová, G. On the Origin of the Functional Versatility of Macrophages. Front. Physiol. 2023, 14, 1128984. [Google Scholar] [CrossRef]
  16. Sreejit, G.; Fleetwood, A.J.; Murphy, A.J.; Nagareddy, P.R. Origins and Diversity of Macrophages in Health and Disease. Clin. Transl. Immunol. 2020, 9, e1222. [Google Scholar] [CrossRef]
  17. Yona, S.; Kim, K.-W.; Wolf, Y.; Mildner, A.; Varol, D.; Strauss-Ayali, D.; Viukov, S.; Guilliams, M.; Misharin, A.; Hume, D.; et al. Fate Mapping Reveals Origins and Dynamics of Monocytes and Tissue Macrophages under Homeostasis. Immunity 2013, 38, 79–91, Erratum in Immunity 2013, 38, 1073–1079.. [Google Scholar] [CrossRef]
  18. Lendeckel, U.; Venz, S.; Wolke, C. Macrophages: Shapes and Functions. Chemtexts 2022, 8, 12. [Google Scholar] [CrossRef]
  19. Viola, M.F.; Franco Taveras, E.; Mass, E. Developmental Programming of Tissue-Resident Macrophages. Front. Immunol. 2024, 15, 1475369. [Google Scholar] [CrossRef]
  20. Honold, L.; Nahrendorf, M. Resident and Monocyte-Derived Macrophages in Cardiovascular Disease. Circ. Res. 2018, 122, 113–127. [Google Scholar] [CrossRef]
  21. Rasheed, A.; Rayner, K.J. Macrophage Responses to Environmental Stimuli During Homeostasis and Disease. Endocr. Rev. 2021, 42, 407–435. [Google Scholar] [CrossRef]
  22. Xue, J.-D.; Gao, J.; Tang, A.-F.; Feng, C. Shaping the Immune Landscape: Multidimensional Environmental Stimuli Refine Macrophage Polarization and Foster Revolutionary Approaches in Tissue Regeneration. Heliyon 2024, 10, e37192. [Google Scholar] [CrossRef] [PubMed]
  23. Murray, P.J.; Wynn, T.A. Protective and Pathogenic Functions of Macrophage Subsets. Nat. Rev. Immunol. 2011, 11, 723–737. [Google Scholar] [CrossRef] [PubMed]
  24. Yunna, C.; Mengru, H.; Lei, W.; Weidong, C. Macrophage M1/M2 Polarization. Eur. J. Pharmacol. 2020, 877, 173090. [Google Scholar] [CrossRef]
  25. Pan, Y.; Yu, Y.; Wang, X.; Zhang, T. Tumor-Associated Macrophages in Tumor Immunity. Front. Immunol. 2020, 11, 583084, Erratum in Front. Immunol. 2021, 12, 775758.. [Google Scholar] [CrossRef] [PubMed]
  26. Hsu, H.-Y.; Wen, M.-H. Lipopolysaccharide-Mediated Reactive Oxygen Species and Signal Transduction in the Regulation of Interleukin-1 Gene Expression. J. Biol. Chem. 2002, 277, 22131–22139, Erratum in J. Biol. Chem. 2002, 277, 33530.. [Google Scholar] [CrossRef]
  27. Ni, R.; Jiang, L.; Zhang, C.; Liu, M.; Luo, Y.; Hu, Z.; Mou, X.; Zhu, Y. Biologic Mechanisms of Macrophage Phenotypes Responding to Infection and the Novel Therapies to Moderate Inflammation. Int. J. Mol. Sci. 2023, 24, 8358. [Google Scholar] [CrossRef]
  28. Cutolo, M.; Soldano, S.; Smith, V.; Gotelli, E.; Hysa, E. Dynamic Macrophage Phenotypes in Autoimmune and Inflammatory Rheumatic Diseases. Nat. Rev. Rheumatol. 2025, 21, 546–565. [Google Scholar] [CrossRef]
  29. Strizova, Z.; Benesova, I.; Bartolini, R.; Novysedlak, R.; Cecrdlova, E.; Foley, L.K.; Striz, I. M1/M2 Macrophages and Their Overlaps—Myth or Reality? Clin. Sci. 2023, 137, 1067–1093. [Google Scholar] [CrossRef]
  30. Tannahill, G.M.; Curtis, A.M.; Adamik, J.; Palsson-McDermott, E.M.; McGettrick, A.F.; Goel, G.; Frezza, C.; Bernard, N.J.; Kelly, B.; Foley, N.H.; et al. Succinate Is an Inflammatory Signal That Induces IL-1β through HIF-1α. Nature 2013, 496, 238–242. [Google Scholar] [CrossRef]
  31. Van den Bossche, J.; Baardman, J.; Otto, N.A.; van der Velden, S.; Neele, A.E.; van den Berg, S.M.; Luque-Martin, R.; Chen, H.-J.; Boshuizen, M.C.S.; Ahmed, M.; et al. Mitochondrial Dysfunction Prevents Repolarization of Inflammatory Macrophages. Cell Rep. 2016, 17, 684–696. [Google Scholar] [CrossRef]
  32. Lu, G.; Zhang, R.; Geng, S.; Peng, L.; Jayaraman, P.; Chen, C.; Xu, F.; Yang, J.; Li, Q.; Zheng, H.; et al. Myeloid Cell-Derived Inducible Nitric Oxide Synthase Suppresses M1 Macrophage Polarization. Nat. Commun. 2015, 6, 6676. [Google Scholar] [CrossRef]
  33. Dousdampanis, P.; Aggeletopoulou, I.; Mouzaki, A. The Role of M1/M2 Macrophage Polarization in the Pathogenesis of Obesity-Related Kidney Disease and Related Pathologies. Front. Immunol. 2024, 15, 1534823. [Google Scholar] [CrossRef]
  34. Peng, Y.; Zhou, M.; Yang, H.; Qu, R.; Qiu, Y.; Hao, J.; Bi, H.; Guo, D. Regulatory Mechanism of M1/M2 Macrophage Polarization in the Development of Autoimmune Diseases. Mediat. Inflamm. 2023, 2023, 8821610. [Google Scholar] [CrossRef] [PubMed]
  35. Rőszer, T. Understanding the Mysterious M2 Macrophage through Activation Markers and Effector Mechanisms. Mediat. Inflamm. 2015, 2015, 816460. [Google Scholar] [CrossRef] [PubMed]
  36. Lyu, L.; Cai, Y.; Zhang, G.; Jing, Z.; Liang, J.; Zhang, R.; Dang, X.; Zhang, C. Exosomes Derived from M2 Macrophages Induce Angiogenesis to Promote Wound Healing. Front. Mol. Biosci. 2022, 9, 1008802. [Google Scholar] [CrossRef] [PubMed]
  37. Chen, C.; Tang, Y.; Zhu, X.; Yang, J.; Liu, Z.; Chen, Y.; Wang, J.; Shang, R.; Zheng, W.; Zhang, X.; et al. P311 Promotes IL-4 Receptor–Mediated M2 Polarization of Macrophages to Enhance Angiogenesis for Efficient Skin Wound Healing. J. Investig. Dermatol. 2023, 143, 648–660.e6. [Google Scholar] [CrossRef]
  38. Viola, A.; Munari, F.; Sánchez-Rodríguez, R.; Scolaro, T.; Castegna, A. The Metabolic Signature of Macrophage Responses. Front. Immunol. 2019, 10, 1462. [Google Scholar] [CrossRef]
  39. Watanabe, S.; Alexander, M.; Misharin, A.V.; Budinger, G.R.S. The Role of Macrophages in the Resolution of Inflammation. J. Clin. Investig. 2019, 129, 2619–2628. [Google Scholar] [CrossRef]
  40. Brancewicz, J.; Wójcik, N.; Sarnowska, Z.; Robak, J.; Król, M. The Multifaceted Role of Macrophages in Biology and Diseases. Int. J. Mol. Sci. 2025, 26, 2107. [Google Scholar] [CrossRef]
  41. Park, M.D.; Silvin, A.; Ginhoux, F.; Merad, M. Macrophages in Health and Disease. Cell 2022, 185, 4259–4279. [Google Scholar] [CrossRef]
  42. Pérez, S.; Rius-Pérez, S. Macrophage Polarization and Reprogramming in Acute Inflammation: A Redox Perspective. Antioxidants 2022, 11, 1394. [Google Scholar] [CrossRef]
  43. Sun, X.; Li, Y.; Deng, Q.; Hu, Y.; Dong, J.; Wang, W.; Wang, Y.; Li, C. Macrophage Polarization, Metabolic Reprogramming, and Inflammatory Effects in Ischemic Heart Disease. Front. Immunol. 2022, 13, 934040. [Google Scholar] [CrossRef] [PubMed]
  44. Zhang, Y.-F.; Shi, T.T.; Lin, Y.-L.; Zhu, Y.-T.; Lin, S.; Fang, T.-Y. Macrophage Metabolic Reprogramming in Inflammatory Bowel Diseases: From Pathogenesis to Therapy. J. Inflamm. Res. 2025, 18, 11821–11839. [Google Scholar] [CrossRef]
  45. Fedotova, E.I.; Berezhnov, A.V.; Popov, D.Y.; Shitikova, E.Y.; Vinokurov, A.Y. The Role of MtDNA Mutations in Atherosclerosis: The Influence of Mitochondrial Dysfunction on Macrophage Polarization. Int. J. Mol. Sci. 2025, 26, 1019. [Google Scholar] [CrossRef] [PubMed]
  46. Khallou-Laschet, J.; Varthaman, A.; Fornasa, G.; Compain, C.; Gaston, A.-T.; Clement, M.; Dussiot, M.; Levillain, O.; Graff-Dubois, S.; Nicoletti, A.; et al. Macrophage Plasticity in Experimental Atherosclerosis. PLoS ONE 2010, 5, e8852. [Google Scholar] [CrossRef]
  47. Laria, A.; Lurati, A.; Marrazza, M.; Mazzocchi, D.; Re, K.A.; Scarpellini, M. The Macrophages in Rheumatic Diseases. J. Inflamm. Res. 2016, 9, 1–11. [Google Scholar] [CrossRef]
  48. Fukui, S.; Iwamoto, N.; Takatani, A.; Igawa, T.; Shimizu, T.; Umeda, M.; Nishino, A.; Horai, Y.; Hirai, Y.; Koga, T.; et al. M1 and M2 Monocytes in Rheumatoid Arthritis: A Contribution of Imbalance of M1/M2 Monocytes to Osteoclastogenesis. Front. Immunol. 2017, 8, 1958. [Google Scholar] [CrossRef] [PubMed]
  49. Kraakman, M.J.; Murphy, A.J.; Jandeleit-Dahm, K.; Kammoun, H.L. Macrophage Polarization in Obesity and Type 2 Diabetes: Weighing down Our Understanding of Macrophage Function? Front. Immunol. 2014, 5, 470. [Google Scholar] [CrossRef]
  50. Fujisaka, S. The Role of Adipose Tissue M1/M2 Macrophages in Type 2 Diabetes Mellitus. Diabetol. Int. 2021, 12, 74–79. [Google Scholar] [CrossRef]
  51. Panda, S.; Arora, A.; Luthra, K.; Mohan, A.; Vikram, N.K.; Kumar Gupta, N.; Singh, A. Hyperglycemia Modulates M1/M2 Macrophage Polarization in Chronic Diabetic Patients with Pulmonary Tuberculosis Infection. Immunobiology 2024, 229, 152787. [Google Scholar] [CrossRef]
  52. Yao, K.; Zhao, Y.-F. Aging Modulates Microglia Phenotypes in Neuroinflammation of MPTP-PD Mice. Exp. Gerontol. 2018, 111, 86–93. [Google Scholar] [CrossRef] [PubMed]
  53. Fang, H.; Chi, X.; Wang, M.; Liu, J.; Sun, M.; Zhang, J.; Zhang, W. M2 Macrophage-Derived Exosomes Promote Cell Proliferation, Migration and EMT of Non-Small Cell Lung Cancer by Secreting MiR-155-5p. Mol. Cell. Biochem. 2025, 480, 3019–3032. [Google Scholar] [CrossRef] [PubMed]
  54. Dang, T.; Liou, G.-Y. Macrophage Cytokines Enhance Cell Proliferation of Normal Prostate Epithelial Cells through Activation of ERK and Akt. Sci. Rep. 2018, 8, 7718. [Google Scholar] [CrossRef] [PubMed]
  55. Tu, D.; Dou, J.; Wang, M.; Zhuang, H.; Zhang, X. M2 Macrophages Contribute to Cell Proliferation and Migration of Breast Cancer. Cell Biol. Int. 2021, 45, 831–838. [Google Scholar] [CrossRef]
  56. Jiang, Y.; Cai, R.; Huang, Y.; Zhu, L.; Xiao, L.; Wang, C.; Wang, L. Macrophages in Organ Fibrosis: From Pathogenesis to Therapeutic Targets. Cell Death Discov. 2024, 10, 487. [Google Scholar] [CrossRef]
  57. Braga, T.T.; Agudelo, J.S.H.; Camara, N.O.S. Macrophages During the Fibrotic Process: M2 as Friend and Foe. Front. Immunol. 2015, 6, 602. [Google Scholar] [CrossRef]
  58. Wang, S.; Wang, J.; Chen, Z.; Luo, J.; Guo, W.; Sun, L.; Lin, L. Targeting M2-like Tumor-Associated Macrophages Is a Potential Therapeutic Approach to Overcome Antitumor Drug Resistance. NPJ Precis. Oncol. 2024, 8, 31. [Google Scholar] [CrossRef]
  59. Liu, J.; Geng, X.; Hou, J.; Wu, G. New Insights into M1/M2 Macrophages: Key Modulators in Cancer Progression. Cancer Cell Int. 2021, 21, 389. [Google Scholar] [CrossRef]
  60. Jin, H.; Meng, X.; Feng, J. Mechanisms of Tumor-Associated Macrophages in Breast Cancer and Treatment Strategy. Front. Immunol. 2025, 16, 1560393. [Google Scholar] [CrossRef]
  61. Ren, X.; Wang, Z.; Wang, J.; Li, X.; Wei, H.; Liu, C.; Liu, S.; Zhu, Y.; Feng, C.; Yin, Y.; et al. The Effects of M2 Macrophages-Derived Exosomes on Urethral Fibrosis and Stricture in Scar Formation. ImmunoTargets Ther. 2025, 14, 151–173. [Google Scholar] [CrossRef]
  62. Chen, R.; Zhang, H.; Tang, B.; Luo, Y.; Yang, Y.; Zhong, X.; Chen, S.; Xu, X.; Huang, S.; Liu, C. Macrophages in Cardiovascular Diseases: Molecular Mechanisms and Therapeutic Targets. Signal Transduct. Target. Ther. 2024, 9, 130. [Google Scholar] [CrossRef]
  63. Li, X.; Liu, Y.; Tang, Y.; Xia, Z. Transformation of Macrophages into Myofibroblasts in Fibrosis-Related Diseases: Emerging Biological Concepts and Potential Mechanism. Front. Immunol. 2024, 15, 1474688. [Google Scholar] [CrossRef]
  64. Chen, B.; Yang, Y.; Yang, C.; Duan, J.; Chen, L.; Lu, K.; Yi, B.; Chen, Y.; Xu, D.; Huang, H. M2 Macrophage Accumulation Contributes to Pulmonary Fibrosis, Vascular Dilatation, and Hypoxemia in Rat Hepatopulmonary Syndrome. J. Cell. Physiol. 2021, 236, 7682–7697. [Google Scholar] [CrossRef]
  65. Hou, J.; Shi, J.; Chen, L.; Lv, Z.; Chen, X.; Cao, H.; Xiang, Z.; Han, X. M2 Macrophages Promote Myofibroblast Differentiation of LR-MSCs and Are Associated with Pulmonary Fibrogenesis. Cell Commun. Signal. 2018, 16, 89. [Google Scholar] [CrossRef]
  66. Hu, M.; Yao, Z.; Xu, L.; Peng, M.; Deng, G.; Liu, L.; Jiang, X.; Cai, X. M2 Macrophage Polarization in Systemic Sclerosis Fibrosis: Pathogenic Mechanisms and Therapeutic Effects. Heliyon 2023, 9, e16206. [Google Scholar] [CrossRef]
  67. Berridge, M.J.; Lipp, P.; Bootman, M.D. The Versatility and Universality of Calcium Signalling. Nat. Rev. Mol. Cell Biol. 2000, 1, 11–21. [Google Scholar] [CrossRef]
  68. Desai, B.N.; Leitinger, N. Purinergic and Calcium Signaling in Macrophage Function and Plasticity. Front. Immunol. 2014, 5, 580. [Google Scholar] [CrossRef] [PubMed]
  69. Demaurex, N.; Nunes, P. The Role of STIM and ORAI Proteins in Phagocytic Immune Cells. Am. J. Physiol. Physiol. 2016, 310, C496–C508. [Google Scholar] [CrossRef] [PubMed]
  70. Beceiro, S.; Radin, J.N.; Chatuvedi, R.; Piazuelo, M.B.; Horvarth, D.J.; Cortado, H.; Gu, Y.; Dixon, B.; Gu, C.; Lange, I.; et al. TRPM2 Ion Channels Regulate Macrophage Polarization and Gastric Inflammation during Helicobacter Pylori Infection. Mucosal Immunol. 2017, 10, 493–507. [Google Scholar] [CrossRef] [PubMed]
  71. Qin, J.; Zhang, X.; Tan, B.; Zhang, S.; Yin, C.; Xue, Q.; Zhang, Z.; Ren, H.; Chen, J.; Liu, M.; et al. Blocking P2X7-Mediated Macrophage Polarization Overcomes Treatment Resistance in Lung Cancer. Cancer Immunol. Res. 2020, 8, 1426–1439. [Google Scholar] [CrossRef]
  72. Marques-da-Silva, C.; Chaves, M.M.; Rodrigues, J.C.; Corte-Real, S.; Coutinho-Silva, R.; Persechini, P.M. Differential Modulation of ATP-Induced P2X7-Associated Permeabilities to Cations and Anions of Macrophages by Infection with Leishmania Amazonensis. PLoS ONE 2011, 6, e25356. [Google Scholar] [CrossRef]
  73. Pelegrin, P.; Surprenant, A. Dynamics of Macrophage Polarization Reveal New Mechanism to Inhibit IL-1beta Release through Pyrophosphates. EMBO J. 2009, 28, 2114–2127. [Google Scholar] [CrossRef]
  74. Layhadi, J.A.; Fountain, S.J. P2X4 Receptor-Dependent Ca(2+) Influx in Model Human Monocytes and Macrophages. Int. J. Mol. Sci. 2017, 18, 2261. [Google Scholar] [CrossRef] [PubMed]
  75. Bkaily, G.; Jacques, D. L-Type Calcium Channel Antagonists and Suppression of Expression of Plasminogen Receptors: Is the Missing Link the L-Type Calcium Channel? Circ. Res. 2009, 105, 112–113. [Google Scholar] [CrossRef]
  76. Molinuevo, M.S.; Etcheverry, S.B.; Cortizo, A.M. Macrophage Activation by a Vanadyl-Aspirin Complex Is Dependent on L-Type Calcium Channel and the Generation of Nitric Oxide. Toxicology 2005, 210, 205–212. [Google Scholar] [CrossRef]
  77. Luciani, D.S.; Gwiazda, K.S.; Yang, T.-L.B.; Kalynyak, T.B.; Bychkivska, Y.; Frey, M.H.Z.; Jeffrey, K.D.; Sampaio, A.V.; Underhill, T.M.; Johnson, J.D. Roles of IP3R and RyR Ca2+ Channels in Endoplasmic Reticulum Stress and Beta-Cell Death. Diabetes 2009, 58, 422–432. [Google Scholar] [CrossRef] [PubMed]
  78. Sermersheim, M.; Kenney, A.D.; Lin, P.-H.; McMichael, T.M.; Cai, C.; Gumpper, K.; Adesanya, T.M.A.; Li, H.; Zhou, X.; Park, K.-H.; et al. MG53 Suppresses Interferon-β and Inflammation via Regulation of Ryanodine Receptor-Mediated Intracellular Calcium Signaling. Nat. Commun. 2020, 11, 3624. [Google Scholar] [CrossRef]
  79. Jacobson, K.A.; Delicado, E.G.; Gachet, C.; Kennedy, C.; von Kügelgen, I.; Li, B.; Miras-Portugal, M.T.; Novak, I.; Schöneberg, T.; Perez-Sen, R.; et al. Update of P2Y Receptor Pharmacology: IUPHAR Review 27. Br. J. Pharmacol. 2020, 177, 2413–2433. [Google Scholar] [CrossRef] [PubMed]
  80. Klaver, D.; Thurnher, M. Control of Macrophage Inflammation by P2Y Purinergic Receptors. Cells 2021, 10, 1098. [Google Scholar] [CrossRef]
  81. Merz, J.; Nettesheim, A.; von Garlen, S.; Albrecht, P.; Saller, B.S.; Engelmann, J.; Hertle, L.; Schäfer, I.; Dimanski, D.; König, S.; et al. Pro- and Anti-Inflammatory Macrophages Express a Sub-Type Specific Purinergic Receptor Profile. Purinergic Signal. 2021, 17, 481–492. [Google Scholar] [CrossRef]
  82. Sakaki, H.; Tsukimoto, M.; Harada, H.; Moriyama, Y.; Kojima, S. Autocrine Regulation of Macrophage Activation via Exocytosis of ATP and Activation of P2Y11 Receptor. PLoS ONE 2013, 8, e59778. [Google Scholar] [CrossRef]
  83. Gopallawa, I.; Freund, J.R.; Lee, R.J. Bitter Taste Receptors Stimulate Phagocytosis in Human Macrophages through Calcium, Nitric Oxide, and Cyclic-GMP Signaling. Cell. Mol. Life Sci. 2021, 78, 271–286. [Google Scholar] [CrossRef] [PubMed]
  84. Seegren, P.V.; Harper, L.R.; Downs, T.K.; Zhao, X.-Y.; Viswanathan, S.B.; Stremska, M.E.; Olson, R.J.; Kennedy, J.; Ewald, S.E.; Kumar, P.; et al. Reduced Mitochondrial Calcium Uptake in Macrophages Is a Major Driver of Inflammaging. Nat. Aging 2023, 3, 796–812. [Google Scholar] [CrossRef] [PubMed]
  85. Okada, H.; Kajiya, H.; Omata, Y.; Matsumoto, T.; Sato, Y.; Kobayashi, T.; Nakamura, S.; Kaneko, Y.; Nakamura, S.; Koyama, T.; et al. CTLA4-Ig Directly Inhibits Osteoclastogenesis by Interfering with Intracellular Calcium Oscillations in Bone Marrow Macrophages. J. Bone Miner. Res. 2019, 34, 1744–1752. [Google Scholar] [CrossRef] [PubMed]
  86. Hanley, P.J.; Musset, B.; Renigunta, V.; Limberg, S.H.; Dalpke, A.H.; Sus, R.; Heeg, K.M.; Preisig-Müller, R.; Daut, J. Extracellular ATP Induces Oscillations of Intracellular Ca2+ and Membrane Potential and Promotes Transcription of IL-6 in Macrophages. Proc. Natl. Acad. Sci. USA 2004, 101, 9479–9484. [Google Scholar] [CrossRef]
  87. Zhao, J.; Zhang, K.; Wang, L.; Zhu, Z.; Jiang, D.; Zuo, Y.; Yang, J.; Jia, W. Macrophage Intracellular “Calcium Oscillations” Triggered Through In Situ Mineralization Regulate Bone Immunity to Facilitate Bone Repair. Adv. Funct. Mater. 2024, 34, 2316224. [Google Scholar] [CrossRef]
  88. Wang, X.; Li, X.; Ong, H.; Tan, T.; Park, K.H.; Bian, Z.; Zou, X.; Haggard, E.; Janssen, P.M.; Merritt, R.E.; et al. MG53 Suppresses NF-ΚB Activation to Mitigate Age-Related Heart Failure. JCI Insight 2021, 6, e148375. [Google Scholar] [CrossRef]
  89. Chen, H.; Ye, H.; Meng, D.-Q.; Cai, P.-C.; Chen, F.; Zhu, L.-P.; Tang, Q.; Long, Z.-X.; Zhou, Q.; Jin, Y.; et al. Reactive Oxygen Species and X-Ray Disrupted Spontaneous [Ca2+]i Oscillation in Alveolar Macrophages. Radiat. Res. 2013, 179, 485–492. [Google Scholar] [CrossRef]
  90. Carrithers, L.M.; Hulseberg, P.; Sandor, M.; Carrithers, M.D. The Human Macrophage Sodium Channel NaV1.5 Regulates Mycobacteria Processing through Organelle Polarization and Localized Calcium Oscillations. FEMS Immunol. Med. Microbiol. 2011, 63, 319–327. [Google Scholar] [CrossRef]
  91. Feske, S.; Wulff, H.; Skolnik, E.Y. Ion Channels in Innate and Adaptive Immunity. Annu. Rev. Immunol. 2015, 33, 291–353. [Google Scholar] [CrossRef]
  92. Feske, S. Calcium Signalling in Lymphocyte Activation and Disease. Nat. Rev. Immunol. 2007, 7, 690–702. [Google Scholar] [CrossRef]
  93. Muik, M.; Schindl, R.; Fahrner, M.; Romanin, C. Ca(2+) Release-Activated Ca(2+) (CRAC) Current, Structure, and Function. Cell. Mol. Life Sci. 2012, 69, 4163–4176. [Google Scholar] [CrossRef] [PubMed]
  94. Feske, S. ORAI1 and STIM1 Deficiency in Human and Mice: Roles of Store-Operated Ca2+ Entry in the Immune System and Beyond. Immunol. Rev. 2009, 231, 189–209. [Google Scholar] [CrossRef]
  95. Ye, Y.; Huang, X.; Zhang, Y.; Lai, X.; Wu, X.; Zeng, X.; Tang, X.; Zeng, Y. Calcium Influx Blocked by SK&F 96365 Modulates the LPS plus IFN-γ-Induced Inflammatory Response in Murine Peritoneal Macrophages. Int. Immunopharmacol. 2012, 12, 384–393. [Google Scholar] [CrossRef]
  96. Nelson, H.A.; Leech, C.A.; Kopp, R.F.; Roe, M.W. Interplay between ER Ca(2+) Binding Proteins, STIM1 and STIM2, Is Required for Store-Operated Ca(2+) Entry. Int. J. Mol. Sci. 2018, 19, 1522. [Google Scholar] [CrossRef]
  97. Chen, Y.-W.; Chen, Y.-F.; Chen, Y.-T.; Chiu, W.-T.; Shen, M.-R. The STIM1-Orai1 Pathway of Store-Operated Ca2+ Entry Controls the Checkpoint in Cell Cycle G1/S Transition. Sci. Rep. 2016, 6, 22142. [Google Scholar] [CrossRef]
  98. Yang, J.; Zhao, Z.; Gu, M.; Feng, X.; Xu, H. Release and Uptake Mechanisms of Vesicular Ca(2+) Stores. Protein Cell 2019, 10, 8–19. [Google Scholar] [CrossRef] [PubMed]
  99. Cao, Q.; Yang, Y.; Zhong, X.Z.; Dong, X.-P. The Lysosomal Ca(2+) Release Channel TRPML1 Regulates Lysosome Size by Activating Calmodulin. J. Biol. Chem. 2017, 292, 8424–8435. [Google Scholar] [CrossRef] [PubMed]
  100. Zajac, M.; Mukherjee, S.; Anees, P.; Oettinger, D.; Henn, K.; Srikumar, J.; Zou, J.; Saminathan, A.; Krishnan, Y. A Mechanism of Lysosomal Calcium Entry. Sci. Adv. 2024, 10, eadk2317. [Google Scholar] [CrossRef]
  101. Kang, H.; Choi, S.W.; Kim, J.Y.; Oh, S.-J.; Kim, S.J.; Lee, M.-S. ER-to-Lysosome Ca(2+) Refilling Followed by K(+) Efflux-Coupled Store-Operated Ca(2+) Entry in Inflammasome Activation and Metabolic Inflammation. eLife 2024, 12, RP87561. [Google Scholar] [CrossRef]
  102. De Marchi, U.; Santo-Domingo, J.; Castelbou, C.; Sekler, I.; Wiederkehr, A.; Demaurex, N. NCLX Protein, but Not LETM1, Mediates Mitochondrial Ca2+ Extrusion, Thereby Limiting Ca2+-Induced NAD(P)H Production and Modulating Matrix Redox State. J. Biol. Chem. 2014, 289, 20377–20385. [Google Scholar] [CrossRef] [PubMed]
  103. dos Santos, G.R.R.M.; Cavalcante Queiroz, M.I.; Leite, A.C.R. LETM1 and Mitochondrial Calcium Homeostasis: A Controversial but Critical Role in Cellular Function and Disease. Cell Calcium 2025, 132, 103085. [Google Scholar] [CrossRef]
  104. Wacquier, B.; Combettes, L.; Dupont, G. Dual Dynamics of Mitochondrial Permeability Transition Pore Opening. Sci. Rep. 2020, 10, 3924. [Google Scholar] [CrossRef]
  105. Kwong, J.Q.; Molkentin, J.D. Physiological and Pathological Roles of the Mitochondrial Permeability Transition Pore in the Heart. Cell Metab. 2015, 21, 206–214. [Google Scholar] [CrossRef] [PubMed]
  106. Yang, X.; Zhuang, J.; Song, W.; Shen, W.; Wu, W.; Shen, H.; Han, S. Mitochondria-Associated Endoplasmic Reticulum Membrane: Overview and Inextricable Link with Cancer. J. Cell. Mol. Med. 2023, 27, 906–919. [Google Scholar] [CrossRef]
  107. Barazzuol, L.; Giamogante, F.; Calì, T. Mitochondria Associated Membranes (MAMs): Architecture and Physiopathological Role. Cell Calcium 2021, 94, 102343. [Google Scholar] [CrossRef]
  108. Missiroli, S.; Patergnani, S.; Caroccia, N.; Pedriali, G.; Perrone, M.; Previati, M.; Wieckowski, M.R.; Giorgi, C. Mitochondria-Associated Membranes (MAMs) and Inflammation. Cell Death Dis. 2018, 9, 329. [Google Scholar] [CrossRef]
  109. Degechisa, S.T.; Dabi, Y.T.; Gizaw, S.T. The Mitochondrial Associated Endoplasmic Reticulum Membranes: A Platform for the Pathogenesis of Inflammation-Mediated Metabolic Diseases. Immunity, Inflamm. Dis. 2022, 10, e647. [Google Scholar] [CrossRef]
  110. Csordás, G.; Renken, C.; Várnai, P.; Walter, L.; Weaver, D.; Buttle, K.F.; Balla, T.; Mannella, C.A.; Hajnóczky, G. Structural and Functional Features and Significance of the Physical Linkage between ER and Mitochondria. J. Cell Biol. 2006, 174, 915–921. [Google Scholar] [CrossRef] [PubMed]
  111. Bernhardt, D.; Müller, M.; Reichert, A.S.; Osiewacz, H.D. Simultaneous Impairment of Mitochondrial Fission and Fusion Reduces Mitophagy and Shortens Replicative Lifespan. Sci. Rep. 2015, 5, 7885. [Google Scholar] [CrossRef] [PubMed]
  112. Rieusset, J. The Role of Endoplasmic Reticulum-Mitochondria Contact Sites in the Control of Glucose Homeostasis: An Update. Cell Death Dis. 2018, 9, 388. [Google Scholar] [CrossRef]
  113. Atakpa-Adaji, P.; Ivanova, A. IP(3)R at ER-Mitochondrial Contact Sites: Beyond the IP(3)R-GRP75-VDAC1 Ca(2+) Funnel. Contact 2023, 6, 25152564231181020. [Google Scholar] [CrossRef]
  114. Rossi, A.; Pizzo, P.; Filadi, R. Calcium, Mitochondria and Cell Metabolism: A Functional Triangle in Bioenergetics. Biochim. Biophys. Acta. Mol. Cell Res. 2019, 1866, 1068–1078. [Google Scholar] [CrossRef]
  115. Baumgartner, H.K.; Gerasimenko, J.V.; Thorne, C.; Ferdek, P.; Pozzan, T.; Tepikin, A.V.; Petersen, O.H.; Sutton, R.; Watson, A.J.M.; Gerasimenko, O. V Calcium Elevation in Mitochondria Is the Main Ca2+ Requirement for Mitochondrial Permeability Transition Pore (MPTP) Opening. J. Biol. Chem. 2009, 284, 20796–20803. [Google Scholar] [CrossRef]
  116. Rottenberg, H.; Hoek, J.B. The Mitochondrial Permeability Transition: Nexus of Aging, Disease and Longevity. Cells 2021, 10, 79. [Google Scholar] [CrossRef] [PubMed]
  117. Elliott, M.R.; Chekeni, F.B.; Trampont, P.C.; Lazarowski, E.R.; Kadl, A.; Walk, S.F.; Park, D.; Woodson, R.I.; Ostankovich, M.; Sharma, P.; et al. Nucleotides Released by Apoptotic Cells Act as a Find-Me Signal to Promote Phagocytic Clearance. Nature 2009, 461, 282–286. [Google Scholar] [CrossRef]
  118. Sawada, K.; Echigo, N.; Juge, N.; Miyaji, T.; Otsuka, M.; Omote, H.; Yamamoto, A.; Moriyama, Y. Identification of a Vesicular Nucleotide Transporter. Proc. Natl. Acad. Sci. USA 2008, 105, 5683–5686. [Google Scholar] [CrossRef]
  119. Di, A.; Krupa, B.; Nelson, D.J. Calcium-G Protein Interactions in the Regulation of Macrophage Secretion. J. Biol. Chem. 2001, 276, 37124–37132. [Google Scholar] [CrossRef]
  120. Zhao, J.; Lu, N.; Qu, Y.; Liu, W.; Zhong, H.; Tang, N.; Li, J.; Wang, L.; Xi, D.; He, F. Calcium-Sensing Receptor-Mediated Macrophage Polarization Improves Myocardial Remodeling in Spontaneously Hypertensive Rats. Exp. Biol. Med. 2024, 249, 10112. [Google Scholar] [CrossRef] [PubMed]
  121. Gutiérrez-López, T.Y.; Orduña-Castillo, L.B.; Hernández-Vásquez, M.N.; Vázquez-Prado, J.; Reyes-Cruz, G. Calcium sensing receptor activates the NLRP3 inflammasome via a chaperone-assisted degradative pathway involving Hsp70 and LC3-II. Biochem. Biophys. Res. Commun. 2018, 505, 1121–1127. [Google Scholar] [CrossRef]
  122. Braun, A.; Gessner, J.E.; Varga-Szabo, D.; Syed, S.N.; Konrad, S.; Stegner, D.; Vögtle, T.; Schmidt, R.E.; Nieswandt, B. STIM1 Is Essential for Fcgamma Receptor Activation and Autoimmune Inflammation. Blood 2009, 113, 1097–1104. [Google Scholar] [CrossRef] [PubMed]
  123. Weber, K.; Schilling, J.D. Lysosomes Integrate Metabolic-Inflammatory Cross-Talk in Primary Macrophage Inflammasome Activation. J. Biol. Chem. 2014, 289, 9158–9171. [Google Scholar] [CrossRef]
  124. Rojanasakul, Y.; Wang, L.; Malanga, C.J.; Ma, J.Y.; Banks, D.E.; Ma, J.K. Altered Calcium Homeostasis and Cell Injury in Silica-Exposed Alveolar Macrophages. J. Cell. Physiol. 1993, 154, 310–316. [Google Scholar] [CrossRef]
  125. Gleva, G.F.; Goodglick, L.A.; Kane, A.B. Altered Calcium Homeostasis in Irreversibly Injured P388D1 Macrophages. Am. J. Pathol. 1990, 137, 43–57. [Google Scholar]
  126. Samie, M.; Wang, X.; Zhang, X.; Goschka, A.; Li, X.; Cheng, X.; Gregg, E.; Azar, M.; Zhuo, Y.; Garrity, A.G.; et al. A TRP Channel in the Lysosome Regulates Large Particle Phagocytosis via Focal Exocytosis. Dev. Cell 2013, 26, 511–524. [Google Scholar] [CrossRef]
  127. Reddy, A.; Caler, E.V.; Andrews, N.W. Plasma Membrane Repair Is Mediated by Ca(2+)-Regulated Exocytosis of Lysosomes. Cell 2001, 106, 157–169. [Google Scholar] [CrossRef]
  128. Nunes, P.; Demaurex, N. The Role of Calcium Signaling in Phagocytosis. J. Leukoc. Biol. 2010, 88, 57–68. [Google Scholar] [CrossRef]
  129. Zimmerli, S.; Majeed, M.; Gustavsson, M.; Stendahl, O.; Sanan, D.A.; Ernst, J.D. Phagosome-Lysosome Fusion Is a Calcium-Independent Event in Macrophages. J. Cell Biol. 1996, 132, 49–61. [Google Scholar] [CrossRef] [PubMed]
  130. Di Virgilio, F.; Meyer, B.C.; Greenberg, S.; Silverstein, S.C. Fc Receptor-Mediated Phagocytosis Occurs in Macrophages at Exceedingly Low Cytosolic Ca2+. J. Cell Biol. 1988, 106, 657–666. [Google Scholar] [CrossRef] [PubMed]
  131. Myers, J.T.; Swanson, J.A. Calcium Spikes in Activated Macrophages during Fcγ Receptor-Mediated Phagocytosis. J. Leukoc. Biol. 2002, 72, 677–684. [Google Scholar] [CrossRef]
  132. Diler, E.; Schwarz, M.; Nickels, R.; Menger, M.D.; Beisswenger, C.; Meier, C.; Tschernig, T. Influence of External Calcium and Thapsigargin on the Uptake of Polystyrene Beads by the Macrophage-like Cell Lines U937 and MH-S. BMC Pharmacol. Toxicol. 2014, 15, 16. [Google Scholar] [CrossRef]
  133. Young, J.D.; Ko, S.S.; Cohn, Z.A. The Increase in Intracellular Free Calcium Associated with IgG Gamma 2b/Gamma 1 Fc Receptor-Ligand Interactions: Role in Phagocytosis. Proc. Natl. Acad. Sci. USA 1984, 81, 5430–5434. [Google Scholar] [CrossRef]
  134. Leon Guerrero, P.A.; Rasmussen, J.P.; Peterman, E. Calcium Dynamics of Skin-Resident Macrophages during Homeostasis and Tissue Injury. Mol. Biol. Cell 2024, 35, br26. [Google Scholar] [CrossRef]
  135. McFadden, M.J.; Reynolds, M.B.; Michmerhuizen, B.C.; Ólafsson, E.B.; Marshall, S.M.; Davis, F.A.; Schultz, T.L.; Iwawaki, T.; Sexton, J.Z.; O’Riordan, M.X.D.; et al. IRE1α Promotes Phagosomal Calcium Flux to Enhance Macrophage Fungicidal Activity. Cell Rep. 2025, 44, 115694. [Google Scholar] [CrossRef] [PubMed]
  136. Zumerle, S.; Calì, B.; Munari, F.; Angioni, R.; Di Virgilio, F.; Molon, B.; Viola, A. Intercellular Calcium Signaling Induced by ATP Potentiates Macrophage Phagocytosis. Cell Rep. 2019, 27, 1–10.e4. [Google Scholar] [CrossRef] [PubMed]
  137. Malik, Z.A.; Denning, G.M.; Kusner, D.J. Inhibition of Ca(2+) Signaling by Mycobacterium Tuberculosis Is Associated with Reduced Phagosome-Lysosome Fusion and Increased Survival within Human Macrophages. J. Exp. Med. 2000, 191, 287–302. [Google Scholar] [CrossRef] [PubMed]
  138. Vashi, N.; Andrabi, S.B.A.; Ghanwat, S.; Suar, M.; Kumar, D. Ca(2+)-Dependent Focal Exocytosis of Golgi-Derived Vesicles Helps Phagocytic Uptake in Macrophages. J. Biol. Chem. 2017, 292, 5144–5165. [Google Scholar] [CrossRef]
  139. Sun, X.; Xu, M.; Cao, Q.; Huang, P.; Zhu, X.; Dong, X.-P. A Lysosomal K(+) Channel Regulates Large Particle Phagocytosis by Facilitating Lysosome Ca(2+) Release. Sci. Rep. 2020, 10, 1038. [Google Scholar] [CrossRef]
  140. Dong, H.; Zhao, B.; Chen, J.; Liu, Z.; Li, X.; Li, L.; Wen, H. Mitochondrial Calcium Uniporter Promotes Phagocytosis-Dependent Activation of the NLRP3 Inflammasome. Proc. Natl. Acad. Sci. USA 2022, 119, e2123247119. [Google Scholar] [CrossRef]
  141. Tedesco, S.; Scattolini, V.; Albiero, M.; Bortolozzi, M.; Avogaro, A.; Cignarella, A.; Fadini, G.P. Mitochondrial Calcium Uptake Is Instrumental to Alternative Macrophage Polarization and Phagocytic Activity. Int. J. Mol. Sci. 2019, 20, 4966. [Google Scholar] [CrossRef]
  142. Murakami, T.; Ockinger, J.; Yu, J.; Byles, V.; McColl, A.; Hofer, A.M.; Horng, T. Critical Role for Calcium Mobilization in Activation of the NLRP3 Inflammasome. Proc. Natl. Acad. Sci. USA 2012, 109, 11282–11287. [Google Scholar] [CrossRef]
  143. Hoffmann, A.; Kann, O.; Ohlemeyer, C.; Hanisch, U.-K.; Kettenmann, H. Elevation of Basal Intracellular Calcium as a Central Element in the Activation of Brain Macrophages (Microglia): Suppression of Receptor-Evoked Calcium Signaling and Control of Release Function. J. Neurosci. 2003, 23, 4410–4419. [Google Scholar] [CrossRef]
  144. Wang, Y.; Li, N.; Zhang, X.; Horng, T. Mitochondrial Metabolism Regulates Macrophage Biology. J. Biol. Chem. 2021, 297, 100904. [Google Scholar] [CrossRef]
  145. Lei, X.; Tan, G.; Wang, Y.; Chen, L.; Cao, Y.; Si, B.; Zhen, Z.; Li, B.; Jin, Y.; Wang, W.; et al. Mitochondrial Calcium Nanoregulators Reverse the Macrophage Proinflammatory Phenotype Through Restoring Mitochondrial Calcium Homeostasis for the Treatment of Osteoarthritis. Int. J. Nanomed. 2023, 18, 1469–1489. [Google Scholar] [CrossRef]
  146. Sipka, T.; Peroceschi, R.; Hassan-Abdi, R.; Groß, M.; Ellett, F.; Begon-Pescia, C.; Gonzalez, C.; Lutfalla, G.; Nguyen-Chi, M. Damage-Induced Calcium Signaling and Reactive Oxygen Species Mediate Macrophage Activation in Zebrafish. Front. Immunol. 2021, 12, 636585. [Google Scholar] [CrossRef]
  147. Nascimento Da Conceicao, V.; Sun, Y.; Zboril, E.K.; De la Chapa, J.J.; Singh, B.B. Loss of Ca(2+) Entry via Orai-TRPC1 Induces ER Stress, Initiating Immune Activation in Macrophages. J. Cell Sci. 2019, 133, jcs237610. [Google Scholar] [CrossRef] [PubMed]
  148. Chauhan, A.; Sun, Y.; Sukumaran, P.; Quenum Zangbede, F.O.; Jondle, C.N.; Sharma, A.; Evans, D.L.; Chauhan, P.; Szlabick, R.E.; Aaland, M.O.; et al. M1 Macrophage Polarization Is Dependent on TRPC1-Mediated Calcium Entry. iScience 2018, 8, 85–102. [Google Scholar] [CrossRef] [PubMed]
  149. Nascimento Da Conceicao, V.; Sun, Y.; Ramachandran, K.; Chauhan, A.; Raveendran, A.; Venkatesan, M.; DeKumar, B.; Maity, S.; Vishnu, N.; Kotsakis, G.A.; et al. Resolving Macrophage Polarization through Distinct Ca(2+) Entry Channel That Maintains Intracellular Signaling and Mitochondrial Bioenergetics. iScience 2021, 24, 103339. [Google Scholar] [CrossRef] [PubMed]
  150. de Azambuja, G.; Moreira Simabuco, F.; Gonçalves de Oliveira, M.C. Macrophage-P2X4 Receptors Pathway Is Essential to Persistent Inflammatory Muscle Hyperalgesia Onset, and Is Prevented by Physical Exercise. PLoS ONE 2025, 20, e0318107. [Google Scholar] [CrossRef]
  151. Wu, C.; Zhao, Y.; Xiao, X.; Fan, Y.; Kloc, M.; Liu, W.; Ghobrial, R.M.; Lan, P.; He, X.; Li, X.C. Graft-Infiltrating Macrophages Adopt an M2 Phenotype and Are Inhibited by Purinergic Receptor P2X7 Antagonist in Chronic Rejection. Am. J. Transplant. 2016, 16, 2563–2573. [Google Scholar] [CrossRef]
  152. Scherr, B.F.; Reiner, M.F.; Baumann, F.; Höhne, K.; Müller, T.; Ayata, K.; Müller-Quernheim, J.; Idzko, M.; Zissel, G. Prevention of M2 Polarization and Temporal Limitation of Differentiation in Monocytes by Extracellular ATP. BMC Immunol. 2023, 24, 11. [Google Scholar] [CrossRef]
  153. de Torre-Minguela, C.; Barberà-Cremades, M.; Gómez, A.I.; Martín-Sánchez, F.; Pelegrín, P. Macrophage Activation and Polarization Modify P2X7 Receptor Secretome Influencing the Inflammatory Process. Sci. Rep. 2016, 6, 22586. [Google Scholar] [CrossRef] [PubMed]
  154. Townsend, E.A.; Guadarrama, A.; Shi, L.; Roti Roti, E.; Denlinger, L.C. P2X(7) Signaling Influences the Production of pro-Resolving and pro-Inflammatory Lipid Mediators in Alveolar Macrophages Derived from Individuals with Asthma. Am. J. Physiol. Lung Cell. Mol. Physiol. 2023, 325, L399–L410. [Google Scholar] [CrossRef] [PubMed]
  155. Chen, K.; Man, Q.; Miao, J.; Xu, W.; Zheng, Y.; Zhou, X.; Gao, Z. Kir2.1 Channel Regulates Macrophage Polarization via the Ca2+/CaMK II/ERK/NF-ΚB Signaling Pathway. J. Cell Sci. 2022, 135, jcs259544. [Google Scholar] [CrossRef] [PubMed]
  156. Murthy, S.; Karkossa, I.; Schmidt, C.; Hoffmann, A.; Hagemann, T.; Rothe, K.; Seifert, O.; Anderegg, U.; von Bergen, M.; Schubert, K.; et al. Danger Signal Extracellular Calcium Initiates Differentiation of Monocytes into SPP1/Osteopontin-Producing Macrophages. Cell Death Dis. 2022, 13, 53. [Google Scholar] [CrossRef]
  157. Liu, H.; Fan, H.; He, P.; Zhuang, H.; Liu, X.; Chen, M.; Zhong, W.; Zhang, Y.; Zhen, C.; Li, Y.; et al. Prohibitin 1 Regulates MtDNA Release and Downstream Inflammatory Responses. EMBO J. 2022, 41, e111173. [Google Scholar] [CrossRef]
  158. Zhang, Y.; Wang, H.; Fu, M.; Yang, L.; Chen, X.; Wang, Z.; Liu, J.; Sun, H. Mechanistic Insights into TSH-Mediated Macrophage Mitochondrial Dysfunction via TSHR Signaling in Metabolic Disorders. Free Radic. Biol. Med. 2025, 240, 783–793. [Google Scholar] [CrossRef]
  159. Ding, Z.; Deng, Y.; Luo, H.; Liu, C.; Yang, M.; Xue, H.; Chen, Z. Progress of Tanshinone IIA against Respiratory Diseases: Therapeutic Targets and Potential Mechanisms. Front. Pharmacol. 2025, 16, 1505672. [Google Scholar] [CrossRef]
  160. Zhou, R.; Yazdi, A.S.; Menu, P.; Tschopp, J. A Role for Mitochondria in NLRP3 Inflammasome Activationc. Nature 2011, 469, 221–225, Erratum in Nature 2011, 475, 122.. [Google Scholar] [CrossRef]
  161. Zhou, Q.; Wang, Y.; Lu, Z.; He, C.; Li, L.; You, M.; Wang, L.; Cao, T.; Zhao, Y.; Li, Q.; et al. Cx43 Acts as a Mitochondrial Calcium Regulator That Promotes Obesity by Inducing the Polarization of Macrophages in Adipose Tissue. Cell. Signal. 2023, 105, 110606. [Google Scholar] [CrossRef]
  162. Assis, L.H.d.P.; Dorighello, G.d.G.; Oliveira, H.C.F.d. Pro-Inflammatory Polarization of Macrophages Is Associated with Reduced Endoplasmic Reticulum-Mitochondria Interaction. Biochem. Biophys. Res. Commun. 2022, 606, 61–67. [Google Scholar] [CrossRef]
  163. Guiducci, C.; Vicari, A.P.; Sangaletti, S.; Trinchieri, G.; Colombo, M.P. Redirecting in Vivo Elicited Tumor Infiltrating Macrophages and Dendritic Cells towards Tumor Rejection. Cancer Res. 2005, 65, 3437–3446. [Google Scholar] [CrossRef]
  164. Hagemann, T.; Lawrence, T.; McNeish, I.; Charles, K.A.; Kulbe, H.; Thompson, R.G.; Robinson, S.C.; Balkwill, F.R. “Re-Educating” Tumor-Associated Macrophages by Targeting NF-KappaB. J. Exp. Med. 2008, 205, 1261–1268. [Google Scholar] [CrossRef]
  165. Sanson, M.; Distel, E.; Fisher, E.A. HDL Induces the Expression of the M2 Macrophage Markers Arginase 1 and Fizz-1 in a STAT6-Dependent Process. PLoS ONE 2013, 8, e74676. [Google Scholar] [CrossRef]
  166. Tentillier, N.; Etzerodt, A.; Olesen, M.N.; Rizalar, F.S.; Jacobsen, J.; Bender, D.; Moestrup, S.K.; Romero-Ramos, M. Anti-Inflammatory Modulation of Microglia via CD163-Targeted Glucocorticoids Protects Dopaminergic Neurons in the 6-OHDA Parkinson’s Disease Model. J. Neurosci. 2016, 36, 9375–9390. [Google Scholar] [CrossRef]
  167. Adib, Y.; Serror, K.; Pinzon, J.A.; Duciel, L.; Delagrange, M.; Ducos, B.; Boccara, D.; Mimoun, M.; Chaouat, M.; Bensussan, A.; et al. In Vitro Modulation of Macrophage Inflammatory and Pro-Repair Properties Essential for Wound Healing by Calcium and Calcium-Alginate Dressings. Cells 2025, 14, 909. [Google Scholar] [CrossRef] [PubMed]
  168. Xiao, L.; Shiwaku, Y.; Hamai, R.; Tsuchiya, K.; Sasaki, K.; Suzuki, O. Macrophage Polarization Related to Crystal Phases of Calcium Phosphate Biomaterials. Int. J. Mol. Sci. 2021, 22, 11252. [Google Scholar] [CrossRef] [PubMed]
  169. Chen, J.; Zhou, Y.; Lin, X.; Li, H. Macrophage Polarization Related to Biomimetic Calcium Phosphate Coatings: A Preliminary Study. Materials 2022, 16, 332. [Google Scholar] [CrossRef] [PubMed]
  170. Jin, C.; Liang, J.; Wu, J.; Han, X.; Zhou, Y.; Li, B.; Sun, W.; Su, J.; Sun, J.; Wan, S.; et al. Temporal Immunomodulatory Hydrogel Regulating the Immune-Osteogenic Cascade for Infected Bone Defects Regeneration. Adv. Mater. 2025, e14419. [Google Scholar] [CrossRef] [PubMed]
  171. Zhao, Q.; Gong, Z.; Wang, J.; Fu, L.; Zhang, J.; Wang, C.; Miron, R.J.; Yuan, Q.; Zhang, Y. A Zinc- and Calcium-Rich Lysosomal Nanoreactor Rescues Monocyte/Macrophage Dysfunction under Sepsis. Adv. Sci. 2023, 10, e2205097. [Google Scholar] [CrossRef]
  172. Kang, H.; Zhang, K.; Wong, D.S.H.; Han, F.; Li, B.; Bian, L. Near-Infrared Light-Controlled Regulation of Intracellular Calcium to Modulate Macrophage Polarization. Biomaterials 2018, 178, 681–696. [Google Scholar] [CrossRef]
  173. Provost, K.A.; Smith, M.; Arold, S.P.; Hava, D.L.; Sethi, S. Calcium Restores the Macrophage Response to Nontypeable Haemophilus Influenzae in Chronic Obstructive Pulmonary Disease. Am. J. Respir. Cell Mol. Biol. 2015, 52, 728–737. [Google Scholar] [CrossRef]
  174. Arfath, Y.; Kotra, T.; Faizan, M.I.; Akhtar, A.; Abdullah, S.T.; Ahmad, T.; Ahmed, Z.; Rayees, S. TRPV4 Facilitates the Reprogramming of Inflamed Macrophages by Regulating IL-10 Production via CREB. Inflamm. Res. 2024, 73, 1687–1697. [Google Scholar] [CrossRef] [PubMed]
  175. Nasrallah, C.M.; Horvath, T.L. Mitochondrial Dynamics in the Central Regulation of Metabolism. Nat. Rev. Endocrinol. 2014, 10, 650–658. [Google Scholar] [CrossRef] [PubMed]
  176. Weinberg, S.E.; Sena, L.A.; Chandel, N.S. Mitochondria in the Regulation of Innate and Adaptive Immunity. Immunity 2015, 42, 406–417. [Google Scholar] [CrossRef] [PubMed]
  177. Wang, S.; Cao, L.; Huang, C.; Wang, J.; Liu, J.; Wang, Y.; Wang, Q.; Zhou, Q.; Zhang, X.; Zhang, D. Amorphous Calcium Zinc Phosphate Promotes Macrophage-Driven Alveolar Bone Regeneration via Modulation of Energy Metabolism and Mitochondrial Homeostasis. Bioact. Mater. 2025, 52, 829–844. [Google Scholar] [CrossRef]
  178. Feno, S.; Munari, F.; Reane, D.V.; Gissi, R.; Hoang, D.-H.; Castegna, A.; Chazaud, B.; Viola, A.; Rizzuto, R.; Raffaello, A. The Dominant-Negative Mitochondrial Calcium Uniporter Subunit MCUb Drives Macrophage Polarization during Skeletal Muscle Regeneration. Sci. Signal. 2021, 14, eabf3838. [Google Scholar] [CrossRef]
  179. Sun, Q.; Shen, X.; Wang, P.; Ma, J.; Sha, W. Targeting Cyclophilin-D by MiR-1281 Protects Human Macrophages from Mycobacterium Tuberculosis-Induced Programmed Necrosis and Apoptosis. Aging 2019, 11, 12661–12673. [Google Scholar] [CrossRef]
  180. Zhang, Q.; Chen, L.; Liu, J.-Y.; Liu, T.; Wang, R.; Wu, X.-Y.; Li, S.-W. MPTP Mediated Ox-MtDNA Release Inducing Macrophage Pyroptosis and Exacerbating MCD-Induced MASH via Promoting the ITPR3/Ca(2+)/NLRP3 Pathway. J. Transl. Med. 2025, 23, 1289. [Google Scholar] [CrossRef]
Figure 1. Key players of macrophage calcium homeostasis. Activation of metabotropic purinergic receptors (P2Y) and the calcium-sensing receptor (CaSR) stimulates phospholipase C–dependent inositol-1,4,5-trisphosphate (IP3) production, leading to Ca2+ release from the endoplasmic reticulum (ER) via IP3 receptors (IP3R). Ca2+ reuptake into the ER is maintained by SERCA pumps, ensuring restoration of basal calcium homeostasis. Store depletion activates stromal interaction molecule 1 (STIM1) and Orai channels, initiating store-operated calcium entry (SOCE). Ca2+ entry is also mediated by TRPC1 and ionotropic receptors purinergic (P2X). Ca2+ influx in mitochondrial matrix occurs mainly via the mitochondrial calcium uniporter (MCU) while efflux is linked with the function of the Na+/Ca2+ exchanger (NCLX) and (leucine zipper–EF hand-containing transmembrane protein 1) LETM1. Mitochondria-associated membranes (MAMs) facilitate ER-to-mitochondria Ca2+ transfer. Lysosomal channels such as P2X4 and TRPML1 further contribute to intracellular Ca2+ dynamics. Black arrows mean the Ca2+ flows, red arrows mean the influence of P2Y receptors and CaSR activation on RyR and IP3R and the effect of STIM on SOCE.
Figure 1. Key players of macrophage calcium homeostasis. Activation of metabotropic purinergic receptors (P2Y) and the calcium-sensing receptor (CaSR) stimulates phospholipase C–dependent inositol-1,4,5-trisphosphate (IP3) production, leading to Ca2+ release from the endoplasmic reticulum (ER) via IP3 receptors (IP3R). Ca2+ reuptake into the ER is maintained by SERCA pumps, ensuring restoration of basal calcium homeostasis. Store depletion activates stromal interaction molecule 1 (STIM1) and Orai channels, initiating store-operated calcium entry (SOCE). Ca2+ entry is also mediated by TRPC1 and ionotropic receptors purinergic (P2X). Ca2+ influx in mitochondrial matrix occurs mainly via the mitochondrial calcium uniporter (MCU) while efflux is linked with the function of the Na+/Ca2+ exchanger (NCLX) and (leucine zipper–EF hand-containing transmembrane protein 1) LETM1. Mitochondria-associated membranes (MAMs) facilitate ER-to-mitochondria Ca2+ transfer. Lysosomal channels such as P2X4 and TRPML1 further contribute to intracellular Ca2+ dynamics. Black arrows mean the Ca2+ flows, red arrows mean the influence of P2Y receptors and CaSR activation on RyR and IP3R and the effect of STIM on SOCE.
Ijms 26 11901 g001
Figure 2. Differences in maintaining calcium homeostasis in M1 and M2 macrophages. Macrophages with pro-inflammatory effects are characterized by higher basal calcium levels compared to M2-type cells, which is associated with a decrease in mitochondrial Ca2+ buffering properties, including decreased mitochondrial calcium uniporter (MCU) expression. High level of calcium in M1 macrophages leads to the release of various inflammatory cytokines. Transient receptor potential canonical 1 (TRPC1) is also necessary for the polarization of macrophages towards the M1 inflammatory phenotype, while Orai channels, as well as purinergic receptors (P2Y1R and P2Y6R) are more strongly expressed in M2 macrophages. Ionotropic receptors (P2X7) on the other hand, reduce the polarization of macrophages towards M2. Black arrows mean the Ca2+ flows, red arrows mean cytokines release.
Figure 2. Differences in maintaining calcium homeostasis in M1 and M2 macrophages. Macrophages with pro-inflammatory effects are characterized by higher basal calcium levels compared to M2-type cells, which is associated with a decrease in mitochondrial Ca2+ buffering properties, including decreased mitochondrial calcium uniporter (MCU) expression. High level of calcium in M1 macrophages leads to the release of various inflammatory cytokines. Transient receptor potential canonical 1 (TRPC1) is also necessary for the polarization of macrophages towards the M1 inflammatory phenotype, while Orai channels, as well as purinergic receptors (P2Y1R and P2Y6R) are more strongly expressed in M2 macrophages. Ionotropic receptors (P2X7) on the other hand, reduce the polarization of macrophages towards M2. Black arrows mean the Ca2+ flows, red arrows mean cytokines release.
Ijms 26 11901 g002
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

Pogonyalova, M.Y.; Popov, D.Y.; Vinokurov, A.Y. Intracellular Calcium as a Regulator of Polarization and Target Reprogramming of Macrophages. Int. J. Mol. Sci. 2025, 26, 11901. https://doi.org/10.3390/ijms262411901

AMA Style

Pogonyalova MY, Popov DY, Vinokurov AY. Intracellular Calcium as a Regulator of Polarization and Target Reprogramming of Macrophages. International Journal of Molecular Sciences. 2025; 26(24):11901. https://doi.org/10.3390/ijms262411901

Chicago/Turabian Style

Pogonyalova, Marina Y., Daniil Y. Popov, and Andrey Y. Vinokurov. 2025. "Intracellular Calcium as a Regulator of Polarization and Target Reprogramming of Macrophages" International Journal of Molecular Sciences 26, no. 24: 11901. https://doi.org/10.3390/ijms262411901

APA Style

Pogonyalova, M. Y., Popov, D. Y., & Vinokurov, A. Y. (2025). Intracellular Calcium as a Regulator of Polarization and Target Reprogramming of Macrophages. International Journal of Molecular Sciences, 26(24), 11901. https://doi.org/10.3390/ijms262411901

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

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop