Skip to main content
Erschienen in: Advances in Therapy 11/2020

Open Access 15.09.2020 | Review

Mast Cells in Diabetes and Diabetic Wound Healing

verfasst von: Jie Dong, Lihong Chen, Ying Zhang, Navin Jayaswal, Ikram Mezghani, Weijie Zhang, Aristidis Veves

Erschienen in: Advances in Therapy | Ausgabe 11/2020

Abstract

Mast cells (MCs) are granulated, immune cells of the myeloid lineage that are present in connective tissues. Apart from their classical role in allergies, MCs also mediate various inflammatory responses due to the nature of their secretory products. They are involved in important physiological and pathophysiological responses related to inflammation, chronic wounds, and autoimmune diseases. There are also indications that MCs are associated with diabetes and its complications. MCs and MC-derived mediators participate in all wound healing stages and are involved in the pathogenesis of non-healing, chronic diabetic foot ulcers (DFUs). More specifically, recent work has shown increased degranulation of skin MCs in human diabetes and diabetic mice, which is associated with impaired wound healing. Furthermore, MC stabilization, either systemic or local at the skin level, improves wound healing in diabetic mice. Understanding the precise role of MCs in wound progression and healing processes can be of critical importance as it can lead to the development of new targeted therapies for diabetic foot ulceration, one of the most devastating complications of diabetes.
Hinweise

Digital Features

To view digital features for this article go to https://​doi.​org/​10.​6084/​m9.​figshare.​12911561.
Key Summary Points
This is a review paper that focuses on the role of mast cells in diabetic wound healing. Skin mast cells are degranulated in diabetes. Stabilization of the mast cells reduces degranulation and promotes wound healing in animal models. This can be a new treatment for diabetic foot ulceration.

Digital Features

This article is published with digital features to facilitate understanding of the article. To view digital features for this article go to https://​doi.​org/​10.​6084/​m9.​figshare.​12911561.

Introduction

More than 140 years have passed since Paul Ehrlich presented his doctoral thesis, “Contribution to The Theory and Practice of Histological Dyes,” in which mast cells (MCs) were first described [1, 2]. A wealth of knowledge has been generated since, and our perception of the role and functions of MCs in our bodies has been remodeled. From being known for their detrimental role in allergic diseases, such as food allergies, asthma, and anaphylaxis, for decades; to now recognized as crucial players in a diverse array of physiological and pathologic functions, including vasodilation, angiogenesis, lymphangiogenesis, pathogen elimination, innate and adaptive immune responses, wound healing, and homeostasis. Moreover, MCs play an important role in many diseases, such as gastrointestinal disorders, diabetes, malignancies, and cardiovascular diseases [36]. However, to this day, the pathophysiological roles of MCs are not well understood. In this review, we summarize the recent perception of MCs and mainly focus on MCs in diabetes and diabetic wound healing.

MCs Origin and Functions

MCs are granular, long-lived, tissue-residing immune cells derived from precursor cells in the bone marrow [7]. The progenitors release into the circulation and reach various organs to differentiate into multiple MC subtypes. These various subtypes have been characterized in both humans and animals on the basis of differences in cell morphology, histochemical properties, protease content in granules, anaphylactic types of degranulation, receptor expression, and functions [810]. MCs can change their phenotype depending on the tissue environment and culturing conditions [11, 12]. Recently, significant heterogeneity in gene expression of tissue-resident MCs has been described by using a comprehensive analysis of the transcriptome of individual anatomically distinct mouse MC populations [13]. MCs are distributed throughout nearly all tissues and are often found in close proximity to epithelia, fibroblasts, blood and lymphatic vessels, and nerves [6, 14]. They are commonly activated by (1) immunoglobulin E (IgE) antigen cross-linking, which results in the degranulation of the cells and the secretion of various pre-formed molecules including histamine, serotonin, tryptase, chymase, and lipid-derived mediators, as well as by (2) IgE-independent non-allergic responses, which results in the release of both pre-formed and newly synthesized MC mediators, including cytokines and chemokines [1517].
MCs are key initiators and modulators of allergic disorders, such as bronchial asthma, allergic rhinitis, urticaria, food allergies, anaphylaxis, atopic dermatitis, and angioedema [4, 18]. As a result of their location, surface receptors, and a wide spectrum of inflammatory and immunomodulatory mediators, MCs are thought to act as the first sentinels in response to metabolic and immunological changes [1922]. Additionally, MCs communicate and interact with many immune and non-immune cells, such as dendritic cells, macrophages, T cells, B cells, fibroblasts, eosinophils, endothelial cells, and epithelial cells. This facilitates their capacity to be involved in regulating the functions of many tissues and organs [23, 24]. There is increasing evidence that MCs play roles in organ development, skin barrier homeostasis, wound healing, angiogenesis, lymphangiogenesis, heart function, autoimmune diseases, and tumor initiation and progression [5, 6, 14, 25].

MCs in Diabetes

Type 1 Diabetes Mellitus

Type 1 diabetes mellitus (T1DM), or insulin-dependent diabetes mellitus, is an autoimmune disease resulting from interactions of genetic, environmental, and immunologic factors that destroy insulin-producing pancreatic β cells in the islets of Langerhans and lead to insulin deficiency [26, 27]. While significant numbers of islet-infiltrating CD8+ cytotoxic T cells and macrophages have been observed in recent-onset T1DM, making them the most prominent infiltrating cell type in islets devoid of insulin-positive cells [28], several studies demonstrated the importance of other islet-infiltrating cell types such as leukocytes and MCs. Though the role of MCs in the pathogenesis of T1DM is not clear, it is generally accepted that MCs are able to present antigens to activate T cells in major histocompatibility complex class I and class II pathways in rodents and humans. It has also been suggested that MCs promote T cell migration into inflammatory sites by producing chemokines, or indirectly by increasing endothelial cell adhesion molecule expression [29]. In samples from donors with T1DM, a larger number of MCs were found to infiltrate pancreatic islets compared with those from donors without diabetes or with type 2 diabetes mellitus (T2DM). Furthermore, the extent of the infiltration correlated with β cell damage. Histamine, which is found at high levels in MCs, directly contributed to β cell death in isolated human islets and INS-1E cells, an insulin-secreting cell line, via a caspase-independent pathway [30]. The observation that patients with T1DM and their siblings have increased levels of circulating IgE compared to the general population also suggests that IgE-mediated activation of MCs could be involved in the pathogenesis of T1DM [31]. However, the exact role of MC, if any, in the activation and migration of autoreactive T cells to the islets has not been demonstrated, raising doubts regarding their possible role.
The DRlyp/lyp rats are a T1DM animal model that develops spontaneous T1DM with hyperglycemia, glycosuria, weight loss, decreased plasma insulin, and peripheral T cell lymphopenia. In contrast, the wild-type DR+/+ rats do not develop diabetes but have a genetic predisposition. Previous studies have shown that in pancreatic lymph nodes (PLN), MCs were more abundant in DRlyp/lyp rats than in DR+/+ rats and that their gene expression profile revealed an upregulation in specific MC genes. When treated with the MCs stabilizer disodium cromoglycate (DSCG or cromolyn), a membrane stabilizer that inhibits MC degranulation, DRlyp/lyp rats show delayed T1DM onset [32]. However, those results were not definitive since it is now clear that cromolyn is not a highly selective MC blocker and can affect other innate immune cells such as macrophages and basophils [33].
The role of MCs in the streptozocin-induced model of T1DM was evaluated by employing W/Wv or Wsh/Wsh MC-deficient mice. MC-deficient mice developed severe insulitis and accelerated hyperglycemia, with 100% of mice becoming diabetic compared to their littermates. They also had decreased numbers of T regulatory cells in the PLNs, and MCs deficiency caused a significant reduction in interleukins (IL), such as IL-10, IL-6, and transforming growth factor-beta (TGFβ) expression in the pancreatic tissue. These results indicate that MCs play a protective role in T1DM and suggest that a failure in MCs expression and function leads to the increased severity of this disease [34]. However, additional studies in two nonobese diabetic (NOD) mouse models with MC deficiency, namely the NOD.kitW-sh/W-sh and NOD.Cpa3Cre/+ mice, showed that the absence of MCs did not affect the progression or incidence of T1DM and had no discernible effects on the overall autoimmune response, which involves both innate and adaptive immune components [35]. Finally, in NOD mice, conditional MCs knockout that results in MCs selective depletion at an early stage, protects the mice from autoimmune T1DM. MCs of NOD mice were overly inflammatory, and they secrete a large amount of IL-6, which favors the differentiation of IL-17-secreting T cells but failed to acquire a tolerogenic IL-10+ phenotype [36]. The controversies of these outcomes partly result from the different models applied. Moreover, some of these models carry various immune abnormalities or have an increase or decrease in other cell types that can affect T1DM [37, 38].

Type 2 Diabetes Mellitus

Type 2 diabetes mellitus (T2DM), or non-insulin-dependent diabetes mellitus, is the most common form of diabetes. It is characterized by increased plasma glucose levels due to insulin secretion deficiencies and insulin resistance, which can be a direct consequence of obesity. Obesity is associated with chronic, low-grade inflammation of adipose tissue (AT), characterized by recruitment of immune cells such as proinflammatory M1 macrophages, neutrophils, natural killer cells, T helper cells type 1, T helper cells type 17, B cells, and MCs [3942]. AT is a reservoir for MCs, where their numbers are found to be particularly increased in obese individuals compared with lean subjects. Most MCs are degranulated as tryptase concentrations are significantly higher in serum obtained from patients with obesity [43, 44]. As potent inducers of inflammatory responses, MCs could potentially contribute to obesity-induced AT inflammation and metabolic dysregulation [45]. MC-deficient KitW-sh/W-sh mice and wild-type mice that are treated with cromolyn to stabilize MCs do not develop obesity in response to high-fat diet, nor do they show signs of increased levels of inflammatory cytokines, chemokines, and proteases in serum and white adipose tissue (WAT). Moreover, proinflammatory MCs play detrimental roles in obesity and diabetes. MCs in WAT obtained from lean humans and mice were leptin-deficient and polarized macrophages toward an anti-inflammatory M2 phenotype. An adoptive transfer of leptin-deficient MCs mitigates obesity and diabetes in obese mice [46].
MCs also play a role in regulating AT thermogenesis. Using the tryptophan hydroxylase 1 (Tph1) inhibitor and Tph1-deficient MCs, studies showed that MC-derived serotonin inhibits subcutaneous adipose tissue browning and systemic energy expenditure. Functional inactivation of MCs or inhibition of MC serotonin synthesis in subcutaneous adipose tissue promotes adipocyte browning and systemic energy metabolism in mice [47]. Furthermore, the exposure of mice to thermoneutrality promotes the infiltration of WAT with MCs that are highly enriched with Tph1. Engraftment of MC-deficient mice with Tph1−/− MCs or selective MCs deletion of Tph1 enhances uncoupling protein 1 expression in WAT and protects mice from developing obesity and insulin resistance [48].
However, conflicting findings have been reported on metabolic dysfunction that is related to obesity in MC-deficient mice, which are presented in detail in Table 1. More specifically, not only did some studies demonstrate that the absence of MCs does not affect AT inflammation and metabolic dysregulation but they also showed the protection of Kit mutant mice against the effects of hypercaloric diet resulted from effects of reduced Kit expression rather than MCs deficiency [49]. Studies on diet-induced obesity in Mcpt5-Cre R-DTA MC-deficient mice, in which the lack of MCs is caused by a principle different from MCs deficiency in Cpa3Cre/+ mice or Kit mutations, also observed no differences in terms of accumulation of M1 macrophages or upregulation of inflammatory cytokines including IL-1β, IL-6, IL-10, and tumor necrosis factor (TNF) between the MC-deficient and MC-proficient mice. Furthermore, MC deficiency had no marked changes in obesity and obesity-related dysregulation observed in weight gain, glucose tolerance, insulin resistance, and other metabolic parameters [50]. In another study, wild-type and MC-deficient mice were fed a high-fat or low-fat diet to study MC influence on inflammatory cell polarization in WAT and overall metabolic changes. The results demonstrated that MCs contributed to the local pro-inflammatory state within WAT in obesity but did not play a primary role in causing insulin resistance [51]. Thus, whether MCs play an essential role in obesity and related pathologies is still under debate. Generally, the MC population in AT is dynamic in nature, showing changes associated with tissue remodeling in obesity. On the basis of the anatomical positions of fat pads such as subcutaneous and epididymal fat, MCs may show different activity levels and distributions [52].
Table 1
Studies examining whether metabolic dysfunction is related to obesity in MC-deficient mice
Study year
Model
Main findings
Comments
2006
Geoffrey et al. [32]
DRlyp/lyp and DR+/+ rats
MCs were more abundant in DRlyp/lyp rats than in DR+/+ rats and that their gene expression profile revealed an upregulation in specific MC genes. When treated with the MCs stabilizer disodium cromoglycate (DSCG or cromolyn), a membrane stabilizer that inhibits MCs degranulation, DRlyp/lyp rats show delayed T1DM onset
Those results were not definitive since it is now clear that cromolyn is not a highly selective MC blocker and can have an effect on other innate immune cells such as macrophages and basophils [33]
2015
Carlos et al. [34]
W/Wv or Wsh/Wsh MC-deficient mice
W/Wv or Wsh/Wsh MC-deficient mice developed severe insulitis and accelerated hyperglycemia with 100% of mice becoming diabetic compared to their littermates. They also had decreased numbers of T regulatory cells in the PLNs and MCs deficiency caused a significant reduction in interleukins (IL), such as IL-10, IL-6, and transforming growth factor-beta (TGFβ) expression in the pancreatic tissue
Additional studies in two nonobese diabetic (NOD) mouse models with MC deficiency, namely the NOD.kitW-sh/W-sh and NOD.Cpa3Cre/+ mice, showed that the absence of MCs did not affect the progression or incidence of T1DM and had no discernible effects on the overall autoimmune response, which involves both innate and adaptive immune components [35]
2015
Zhou et al. [46]
MC-deficient KitW-sh/W-sh mice and wild-type mice
MC-deficient KitW-sh/W-sh mice and wild-type mice that are treated with cromolyn to stabilize MCs do not develop obesity in response to high-fat diet nor do they show signs of increased levels of inflammatory cytokines, chemokines, and proteases in serum and white adipose tissue (WAT). Moreover, proinflammatory MCs play detrimental roles in obesity and diabetes. MCs in WAT obtained from lean humans and mice were leptin-deficient and polarized macrophages toward an anti-inflammatory M2 phenotype. An adoptive transfer of leptin-deficient MCs mitigates obesity and diabetes in obese mice
The protection of Kit mutant mice against the effects of hypercaloric diet resulted from effects of reduced Kit expression rather than MCs deficiency [49]
Studies on diet-induced obesity in Mcpt5-Cre R-DTA MC-deficient mice, in which the lack of MCs is caused by a principle different from MCs deficiency in Cpa3Cre/+ mice or Kit mutations, also observed no differences in terms of accumulation of M1 macrophages or upregulation of inflammatory cytokines [50]
In addition to the above, MCs contribute to the progression of complications in T2DM. An increased number of MCs were observed in the kidneys of diabetic animals where they have been found to release various mediators, such as TGFβ, chymase, tryptase, cathepsin G, rennin, and many others. The release of such mediators from MCs may contribute to renal diseases and renal interstitial fibrosis and cause extracellular matrix (ECM) accumulation [53, 54]. A recent study indicated that MC infiltration might promote renal interstitial fibrosis via the stem cell factor (SCF)/c-KIT signaling pathway, which can be inhibited by an antiallergic drug such as tranilast [55]. Another pathway in which MCs contribute to the pathogenesis of diabetic nephropathies might involve the renin–angiotensin system (RAS). Drugs that can inhibit renin directly prevented the increase of MCs in diabetic mice. This was observed when mice were treated with aliskiren [56]. Furthermore, MCs play a substantial role in the development of atherosclerosis in patients with diabetes as they secrete several cytokines including IL-6 and IFNγ, as well as chemokines such as eotaxin, monocyte chemoattractant protein 1 (MCP-1), and RANTES, which are involved in the recruitment of monocytes into the inflammation sites and promote differentiation in the arterial wall. Additionally, MCs participate in lipid retention, vascular cell remodeling, and atherosclerotic plaque progression by releasing vasoactive and angiogenic compounds, and pro-inflammatory mediators, such as arachidonic acid metabolites, histamine, cytokines, chemokines, platelet-activating factors, and proteolytic enzymes [57]. Studies also depicted the contribution of MCs to the pathogenesis of hyperglycemia-induced atrial fibrillation via the enhancement of inflammation and fibrosis with MC-deficient W/W(v) mice [58]. Cardiac MCs were supposed to be activated by metabolic byproducts resulted from hyperglycermia and then participate in the remodeling process of cardiomyopathy by releasing a multitude of cytokines and bioactive enzymes. Increased numbers of MCs and mediator release have been reported in explanted human hearts with dilated cardiomyopathy [59, 60]. Other studies have reported increased cardiac MC density and elevated MC secretions that correlated with gene expression and aberrant cytokine levels associated with cardiac remodeling. Nedocromil, a pharmacologic stabilizer of MC, halted contractile dysfunction in diabetic mice, reduced cardiac MC density, reduced the expression of MMP-2, MMP-9, TNFα, and IFNγ, and increased the expression of IL-4 and IL-10, resulting in attenuation of the diabetes-induced cardiomyopathy [61]. MC-deficiency protects mice from streptozotocin-induced diabetic cardiomyopathy [62]. The main MCs functions are summarized in Fig. 1.

MCs in Wound Healing

Wound healing is a complex and dynamic process that involves coordinated and overlapping phases, including coagulation, inflammation, proliferation, and remodeling aimed at ultimately restoring the barrier function and mechanical integrity of the skin. The vascular system and numerous cell types, cytokines, and mediators are involved in this process [63]. It is initiated with the vasoconstriction of blood vessels and platelet aggregation to stop the bleeding, followed by a flux of a variety of inflammatory cells, such as MCs, neutrophils, macrophages, and endothelial cells. These inflammatory cells then release various mediators and cytokines to promote fibroblast proliferation and migration to the wound area to secrete new ECM and granulation tissue, which then fills the wound prior to re-epithelialization. In the remodeling phase, collagen bundles increase in diameter and become organized to achieve greater tensile strength. Furthermore, fibroblasts undergo a transformation and differentiate into myofibroblasts to facilitate wound contraction [64].
MCs represent up to 8% of the total number of cells within the dermis and are localized adjacent to the epidermis and the subdermal vasculature and nerves. There is evidence supporting the participation of MCs in wound healing owing to their interaction with macrophages, endothelial cells, and fibroblasts [6568]. At the onset of a cutaneous injury, MCs are recruited by anaphylatoxins C3a and C5a, produced under the influence of Hageman factor (XII) and SCF secreted by keratinocytes. Subsequently, recruited MCs release TNFα, histamine, vascular endothelial growth factor (VEGF), IL-6, IL-8, platelet-derived growth factor (PDGF), TGFβ, nerve growth factor, and other mediators. These MC-released mediators play significant roles in the wound healing process, where they (1) contribute to clot stabilization, neoangiogenesis, fibrinogenesis, and re-epithelialization, (2) increase endothelial permeability and vasodilation, (3) facilitate migration of monocytes and neutrophils to the site of injury, and (4) activate keratinocytes and fibroblasts. It has been observed that early skin wound healing is impaired in the absence of MCs, and after wound healing, vascular permeability and neutrophil recruitment is decreased. MCs degranulate in response to skin wounding and inhibition of histamine, but not in the absence of TNFα, which results in a delayed skin wound closure [67]. MCs were found contributing to scar formation during fetal wound healing. MCs enhance scar formation and mediate the transition from scarless to fibrotic healing during fetal wound repair. MC-deficient (KitW/W-v) was shown to produce less scar tissue when compared to wild-type (Kit+/+) fetuses [69]. The growth and differentiation factor activin A is a key regulator of tissue repair, inflammation, and fibrosis and can increase the number of mature MCs in mouse skin in vivo [69].
MCs were also involved in regulating the homeostatic expression of epidermal differentiation complex (EDC) genes, where decreased barrier functions were found in the absence of MCs. In MC-deficient mice, there is a diminished expression of multiple EDC genes and evidence of increased barrier permeability to protein antigens [70]. MCs have been reported as being more efficient in their ability to downregulate factor XIIIa than in contributing to its amounts and functions in homeostatic conditions. Activated MCs controlled thrombin-induced skin inflammation by MC protease 4, suggesting the complex function of MCs in wound healing, especially in chronic wound healing [71]. Further studies found that MC-deficient mice have a significantly delayed wound closure in infected skin wounds. The delay was associated with impaired bacterial clearance in the absence of MCs [22].
It should be emphasized that there is no universal consensus regarding the role of MCs in wound healing. Thus, studies using mated activin transgenic mice with CreMaster mice observed that loss of MCs did not affect the wound healing process. Furthermore, MC deficiency did not alter wounding-induced inflammation and new tissue formation or chemically induced angiogenesis in mice with normal activin levels. This led to the hypothesis that MCs are not major targets of activin during wound healing and do not play a major role in wound healing in general [72]. However, a drawback of these studies is that they were performed with different mice models, which have additional abnormalities in the immune system, and this can be the main reason for the observed contradictory results. A more specific and reliable model of MC deficiency is needed to fully comprehend the role of MC in normal wound healing.

Diabetic Foot Ulceration

Diabetic foot ulcers (DFUs) are a leading cause of hospital admission for patients with diabetes. DFUs are a major morbidity, often causing patients to suffer from severe pain and a poor quality of life. Approximately 10–15% of patients with diabetes develop foot ulcers, and 15% of patients with DFU require amputation, both of which are associated with high mortality rates of 16.7% at 12 months and over 50% at 5 years [73]. In contrast to non-diabetic wounds, diabetic wounds are characterized by prolonged inflammation, impaired angiogenesis, and delayed wound closure. The etiology of non-healing DFUs is multifactorial because of a combination of peripheral neuropathy, peripheral artery disease, and altered immune function. Additionally, the recruitment of endothelial progenitor cells is impaired in diabetes as a result of reduced nitric oxide production, which ultimately results in impaired angiogenesis. However, the most direct effects on wound healing come from the functional alterations in cells activated by the immune response, including platelets, macrophages, neutrophils, endothelial cells, fibroblasts, and keratinocytes, all contributing to a failure to progress through the normal phases of wound healing [74].
We previously showed that increased skin inflammation and aberrant growth factor levels are the main factors associated with a failure to heal DFUs. Patients with DFUs had more severe neuropathy, higher white blood cell count, and lower endothelium-dependent and independent vasodilation in the macrocirculation. Patients whose ulcers failed to heal had higher TNFα, MCP-1, matrix metallopeptidase-9 (MMP-9), and fibroblast growth factor 2 serum levels when compared with those who healed. Skin biopsy analysis showed that compared with control subjects, patients with diabetes had increased immune cell infiltration, expression of MMP-9, and protein tyrosine phosphatase 1B, which negatively regulates the signaling of insulin, leptin, and various growth factors [75]. We also observed that increased chronic inflammation and blood vessel density in the skin of patients with diabetes and in experimental diabetes models [75, 76], as well as the defective inflammation resolution in diabetic mouse wounds, impair the healing process [77].
Subsequent studies in our unit showed that the number of degranulated MCs was increased in unwounded forearm and foot skin of patients with diabetes and in the unwounded dorsal skin of diabetic mice (Figs. 2 and 3). Conversely, post-wounding MCs degranulation increased in non-diabetic mice, but not in diabetic mice. Pretreatment with the MCs degranulation inhibitor DSCG rescued diabetes-associated wound-healing impairment in mice. Pretreatment with DSCG also shifted macrophages to the regenerative M2 phenotype. Macrophages are critical for the healing of DFUs and can be broadly characterized as “pro-inflammatory” M1 or “immunomodulatory/regenerative” M2 [78, 79]. M1 activation is required during the acute inflammatory phase, although it is also present in chronic wounds characterized by persistent inflammation [80]; whereas M2 activation during the proliferative phase promotes angiogenesis and collagen production [81]. Single-cell RNA sequencing analyses of healthy lower extremity skin and DFUs found that the DFU groups had significantly more M1 polarized as compared to M2 [82]. Furthermore, our studies showed that non-diabetic and diabetic mice deficient in MCs had delayed wound healing compared with their wild-type (WT) controls. These results indicate that intact, non-degranulated MCs are necessary for proper healing, and therapies inhibiting MC degranulation could improve wound healing in diabetes [83].
MCs may also be involved in modulating VEGF, which acts on multiple components of the wound healing cascade, including angiogenesis, epithelization, and collagen deposition. MCs are a significant source of VEGF in mouse skin, and its release from human MCs is reduced in hyperglycemic conditions [84]. Local MC activation increased blood flow through the hind limb (46% at day 9) compared to that in non-activated control mice. Histological analysis of the muscle tissue revealed an increased number of CD31+ capillaries [85].
In addition to the above, additional modes of action for MCs have been proposed. MCs have been shown to promote mesenchymal stem cell (MSC) proliferation and migration by the activation of the PDGF pathway, downregulation of miR-145/143, and modulation of the myocardin–Klf4 axis [86]. Chymase released by MC increased proliferation of skin fibroblast and expression of TGFβ1 and interleukin IL-1β in a dose-dependent way [87]. MC protease mMCP-6, an MC-specific tryptase, has scar-suppressing properties after spinal cord injury via indirect cleavage of axon growth-inhibitory scar components and alteration of the gene expression profile of these factors [88]. MCs protect from the exacerbated allergic skin inflammation induced by repeated allergen challenges [89]. MCs were also utilized as an alternative target in the vasoplegic response in cardiac surgery [90], while MC protease 7 could promote angiogenesis by the degradation of integrin subunits [91].
DFUs treatment requires fully identifying the etiology and assessing the co-morbidities. Adequate care for DFUs should include a focus on appropriate wound debridement, infection control, relieving pressure while standing or walking, and optimizing blood flow. With technological advancement, a series of advanced therapies have been implemented, such as bioengineered skin substitutes, negative pressure wound therapy, and the development of new wound dressings [92]. Though these treatments have provided encouraging results, most of them are expensive, and some even have significant side effects that often result in non-compliance. As MC and their secreted factors can regulate the process of wound healing at multiple levels, MCs stabilizers may be a new therapeutic modality to treat DFUs.

MCs Stabilizers

MC stabilizer ketotifen was used to treat both Yorkshire and red Duroc pigs. Studies showed that ketotifen treatment significantly reduced the first phase of contraction in red Duroc pig wounds to a level equivalent to Yorkshire wounds, but had no detectable effects on the post-epithelialization phase of contraction. Ketotifen treatment also reduced the deposition of collagen within the red Duroc wounds but did not affect Yorkshire pig wounds contraction or collagen deposition, suggesting that ketotifen was an effective treatment for the reduction of excessive wound contraction and fibrosis in human cutaneous injuries without affecting the normal healing process [93]. Another study investigated the effects of systemic usage of DSCG, the most widely used MC stabilizer, on wound healing in BALB/c mice. The authors reported that the application of DSCG could reduce the levels of inflammatory cytokines, such as IL-1α, IL-1β, and CXCL1, decrease scar width, and accelerate collagen reorganization [94].
As our previous studies have shown an increase in degranulated skin MCs in patients with diabetes and animal models of diabetes, a study from our unit investigated the effect of systemic administration of DSCG. It showed that it increased angiogenesis and improved wound healing in diabetic mice [83]. As DSCG may not be a good candidate for topical use, we further developed a new indole carboxamide-type of MCs stabilizer, MCS-01, and proved it to be an effective MCs degranulation inhibitor in vitro by calcium channel blockade. MCS-01 can be delivered topically for prolonged periods through controlled release by specifically designed alginate bandages (Fig. 4). Analyses of MC supernatants showed that MCS-01 significantly inhibited the release of β-hex and TNFα in a concentration-dependent manner. Human LAD2 cells stimulated with substance P (SP, 2 mM), an established inducer of MCs degranulation, showed that MCS-01 inhibited β-hex release and reduced both TNFα and IL-8. This proved that MCS-01 could affect the degranulation and release of pro-inflammatory mediators from MCs. Similar to the effects of a systemic application of DSCG for MCs stabilization, bandages releasing MCS-01 reduced MC degranulation in diabetic mouse skin, and the topical application of MCS-01 accelerated wound healing in both pre- and post-wounding of diabetic mice (Fig. 5). We further investigated the global changes in gene expression patterns. Bulk transcriptome analysis from wounds treated with MCS-01 or placebo showed that MCS-01 (1) significantly modulated messenger RNA and microRNA profiles of diabetic wounds, (2) stimulated upregulation of pathways linked to acute inflammation and immune cell migration, and (3) activated NF-κB, IL-6, and TREM1 signaling pathways. Single-cell RNA sequencing analysis of 6154 cells from wounded and unwounded mouse skin revealed that MCS-01 primarily altered the gene expression of MCs, monocytes, and keratinocytes, while fibroblasts, adipocytes, and T cells had no significant changes. All the above suggests that topical MC stabilization can be a potentially successful treatment for DFUs [95].
Finally, photoimmodulation, or photoimmodulation combined with condition medium derived from human bone marrow MSCs, significantly decreases both the total numbers of MCs and their degranulation and improves wound healing in diabetes [96]. Omega-3 fatty acids significantly decreased the diabetic wound area by affecting the concentration of MCs [97]. Topical administration of 0.03% naltrexone, an opioid receptor antagonist, could accelerate diabetic wound closure by promoting deoxyribonucleic acid synthesis, increasing MCs, and enhancing the expression of PDGF and VEGF [98]. DSCG in subcutaneously mesh-implanted C57BL/6J mice decreased early inflammation and fibrotic responses [99].

Concluding Remarks

Recent work has significantly increased our knowledge of MC as multifaceted immune modulators and operators of health. MCs have also been implicated in the development of non-healing, chronic DFU. The development of new, potent mast cell stabilizers that are appropriate for topical use has the potential to improve impaired diabetic wound healing, one of the most severe complications of diabetes.

Acknowledgements

Funding

This work was supported by the National Institutes of Health Grant DP3DK108224 (A.V.). A.V. received funding from the National Rongxiang Xu Foundation. No Rapid Service or Open Access Fees were received by the journal for the publication of this article.

Authorship

All named authors meet the International Committee of Medical Journal Editors (ICMJE) criteria for authorship for this article, take responsibility for the integrity of the work as a whole, and have given their approval for this version to be published.

Disclosures

Aristidis Veves is a Section Editor for Advances in Therapy. The remaining authors Jie Dong, Lihong Chen, Ying Zhang, Navin Jayaswal, Ikram Mezghani and Weijie Zhang have nothing to disclose.

Compliance with Ethics Guidelines

This article is based on previously conducted studies and does not contain any studies with human participants or animals performed by any of the authors.
Open AccessThis article is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License, which permits any non-commercial use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by-nc/​4.​0/​.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Literatur
2.
Zurück zum Zitat Crivellato E, Beltrami CA, Mallardi F, Ribatti D. Paul Ehrlich’s doctoral thesis: a milestone in the study of mast cells. Br J Haematol. 2003;123:19–21.PubMed Crivellato E, Beltrami CA, Mallardi F, Ribatti D. Paul Ehrlich’s doctoral thesis: a milestone in the study of mast cells. Br J Haematol. 2003;123:19–21.PubMed
3.
Zurück zum Zitat Lyons DO, Pullen NA. Beyond IgE: alternative mast cell activation across different disease states. Int J Mol Sci. 2020;21:1498.PubMedCentral Lyons DO, Pullen NA. Beyond IgE: alternative mast cell activation across different disease states. Int J Mol Sci. 2020;21:1498.PubMedCentral
4.
Zurück zum Zitat Maurer M, Taube C, Schröder NW, et al. Mast cells drive IgE-mediated disease but might be bystanders in many other inflammatory and neoplastic conditions. J Allergy Clin Immunol. 2019;144:S19–30.PubMed Maurer M, Taube C, Schröder NW, et al. Mast cells drive IgE-mediated disease but might be bystanders in many other inflammatory and neoplastic conditions. J Allergy Clin Immunol. 2019;144:S19–30.PubMed
5.
Zurück zum Zitat Varricchi G, Marone G. Mast cells: fascinating but still elusive after 140 years from their discovery. Int J Mol Sci. 2020;21:464. Varricchi G, Marone G. Mast cells: fascinating but still elusive after 140 years from their discovery. Int J Mol Sci. 2020;21:464.
6.
Zurück zum Zitat Varricchi G, Rossi FW, Galdiero MR, et al. Physiological roles of mast cells: Collegium Internationale Allergologicum update 2019. Int Arch Allergy Immunol. 2019;179:247–61.PubMed Varricchi G, Rossi FW, Galdiero MR, et al. Physiological roles of mast cells: Collegium Internationale Allergologicum update 2019. Int Arch Allergy Immunol. 2019;179:247–61.PubMed
7.
Zurück zum Zitat Kirshenbaum AS, Goff JP, Semere T, Foster B, Scott LM, Metcalfe DD. Demonstration that human mast cells arise from a progenitor cell population that is CD34+, c-kit+, and expresses aminopeptidase N (CD13). Blood J Am Soc Hematol. 1999;94:2333–42. Kirshenbaum AS, Goff JP, Semere T, Foster B, Scott LM, Metcalfe DD. Demonstration that human mast cells arise from a progenitor cell population that is CD34+, c-kit+, and expresses aminopeptidase N (CD13). Blood J Am Soc Hematol. 1999;94:2333–42.
8.
Zurück zum Zitat Dahlin JS, Hallgren J. Mast cell progenitors: origin, development and migration to tissues. Mol Immunol. 2015;63:9–17.PubMed Dahlin JS, Hallgren J. Mast cell progenitors: origin, development and migration to tissues. Mol Immunol. 2015;63:9–17.PubMed
9.
Zurück zum Zitat Dvorak AM, Morgan ES. Diamine oxidase-gold enzyme-affinity ultrastructural demonstration that human gut mucosal mast cells secrete histamine by piecemeal degranulation in vivo. J Allergy Clin Immunol. 1997;99:812–20.PubMed Dvorak AM, Morgan ES. Diamine oxidase-gold enzyme-affinity ultrastructural demonstration that human gut mucosal mast cells secrete histamine by piecemeal degranulation in vivo. J Allergy Clin Immunol. 1997;99:812–20.PubMed
10.
Zurück zum Zitat Welle M. Development, significance, and heterogeneity of mast cells with particular regard to the mast cell-specific proteases chymase and tryptase. J Leukoc Biol. 1997;61:233–45.PubMed Welle M. Development, significance, and heterogeneity of mast cells with particular regard to the mast cell-specific proteases chymase and tryptase. J Leukoc Biol. 1997;61:233–45.PubMed
11.
Zurück zum Zitat Bischoff S, Sellge G, Lorentz A, Sebald W, Raab R, Manns M. IL-4 enhances proliferation and mediator release in mature human mast cells. Proc Natl Acad Sci. 1999;96:8080–5.PubMed Bischoff S, Sellge G, Lorentz A, Sebald W, Raab R, Manns M. IL-4 enhances proliferation and mediator release in mature human mast cells. Proc Natl Acad Sci. 1999;96:8080–5.PubMed
12.
Zurück zum Zitat Huber M, Cato AC, Ainooson GK, et al. Regulation of the pleiotropic effects of tissue-resident mast cells. J Allergy Clin Immunol. 2019;144:S31–45.PubMed Huber M, Cato AC, Ainooson GK, et al. Regulation of the pleiotropic effects of tissue-resident mast cells. J Allergy Clin Immunol. 2019;144:S31–45.PubMed
13.
Zurück zum Zitat Dwyer DF, Barrett NA, Austen KF. Immunological Genome Project C: expression profiling of constitutive mast cells reveals a unique identity within the immune system. Nat Immunol. 2016;17:878–87.PubMedPubMedCentral Dwyer DF, Barrett NA, Austen KF. Immunological Genome Project C: expression profiling of constitutive mast cells reveals a unique identity within the immune system. Nat Immunol. 2016;17:878–87.PubMedPubMedCentral
14.
Zurück zum Zitat Krystel-Whittemore M, Dileepan KN, Wood JG. Mast cell: a multi-functional master cell. Front Immunol. 2016;6:620.PubMedPubMedCentral Krystel-Whittemore M, Dileepan KN, Wood JG. Mast cell: a multi-functional master cell. Front Immunol. 2016;6:620.PubMedPubMedCentral
17.
Zurück zum Zitat Mukai K, Tsai M, Saito H, Galli SJ. Mast cells as sources of cytokines, chemokines, and growth factors. Immunol Rev. 2018;282:121–50.PubMedPubMedCentral Mukai K, Tsai M, Saito H, Galli SJ. Mast cells as sources of cytokines, chemokines, and growth factors. Immunol Rev. 2018;282:121–50.PubMedPubMedCentral
18.
Zurück zum Zitat Gould HJ, Sutton BJ. IgE in allergy and asthma today. Nat Rev Immunol. 2008;8:205–17.PubMed Gould HJ, Sutton BJ. IgE in allergy and asthma today. Nat Rev Immunol. 2008;8:205–17.PubMed
19.
20.
Zurück zum Zitat Dudeck A, Köberle M, Goldmann O, et al. Mast cells as protectors of health. J Allergy Clin Immunol. 2019;144:S4–18.PubMed Dudeck A, Köberle M, Goldmann O, et al. Mast cells as protectors of health. J Allergy Clin Immunol. 2019;144:S4–18.PubMed
21.
Zurück zum Zitat Piliponsky AM, Acharya M, Shubin NJ. Mast cells in viral, bacterial, and fungal infection immunity. Int J Mol Sci. 2019;20:2851.PubMedCentral Piliponsky AM, Acharya M, Shubin NJ. Mast cells in viral, bacterial, and fungal infection immunity. Int J Mol Sci. 2019;20:2851.PubMedCentral
22.
Zurück zum Zitat Zimmermann C, Troeltzsch D, Giménez-Rivera V, et al. Mast cells are critical for controlling the bacterial burden and the healing of infected wounds. Proc Natl Acad Sci. 2019;116:20500–4.PubMed Zimmermann C, Troeltzsch D, Giménez-Rivera V, et al. Mast cells are critical for controlling the bacterial burden and the healing of infected wounds. Proc Natl Acad Sci. 2019;116:20500–4.PubMed
23.
Zurück zum Zitat Stassen M, Hartmann A-K, Delgado SJ, Dehmel S, Braun A. Mast cells within cellular networks. J Allergy Clin Immunol. 2019;144:S46–54.PubMed Stassen M, Hartmann A-K, Delgado SJ, Dehmel S, Braun A. Mast cells within cellular networks. J Allergy Clin Immunol. 2019;144:S46–54.PubMed
24.
Zurück zum Zitat Gri G, Frossi B, D’Inca F, et al. Mast cell: an emerging partner in immune interaction. Front Immunol. 2012;3:120.PubMedPubMedCentral Gri G, Frossi B, D’Inca F, et al. Mast cell: an emerging partner in immune interaction. Front Immunol. 2012;3:120.PubMedPubMedCentral
25.
Zurück zum Zitat Elieh A, Komi D, Wohrl S, Bielory L. Mast cell biology at molecular level: a comprehensive review. Clin Rev Allergy Immunol. 2020;58:342–65. Elieh A, Komi D, Wohrl S, Bielory L. Mast cell biology at molecular level: a comprehensive review. Clin Rev Allergy Immunol. 2020;58:342–65.
26.
Zurück zum Zitat Bluestone JA, Herold K, Eisenbarth G. Genetics, pathogenesis and clinical interventions in type 1 diabetes. Nature. 2010;464:1293–300.PubMedPubMedCentral Bluestone JA, Herold K, Eisenbarth G. Genetics, pathogenesis and clinical interventions in type 1 diabetes. Nature. 2010;464:1293–300.PubMedPubMedCentral
27.
Zurück zum Zitat Eizirik DL, Colli ML, Ortis F. The role of inflammation in insulitis and β-cell loss in type 1 diabetes. Nature Rev Endocrinol. 2009;5:219. Eizirik DL, Colli ML, Ortis F. The role of inflammation in insulitis and β-cell loss in type 1 diabetes. Nature Rev Endocrinol. 2009;5:219.
28.
Zurück zum Zitat Willcox A, Richardson S, Bone A, Foulis A, Morgan N. Analysis of islet inflammation in human type 1 diabetes. Clin Exp Immunol. 2009;155:173–81.PubMedPubMedCentral Willcox A, Richardson S, Bone A, Foulis A, Morgan N. Analysis of islet inflammation in human type 1 diabetes. Clin Exp Immunol. 2009;155:173–81.PubMedPubMedCentral
29.
Zurück zum Zitat Shi MA, Shi GP. Different roles of mast cells in obesity and diabetes: lessons from experimental animals and humans. Front Immunol. 2012;3:7.PubMedPubMedCentral Shi MA, Shi GP. Different roles of mast cells in obesity and diabetes: lessons from experimental animals and humans. Front Immunol. 2012;3:7.PubMedPubMedCentral
30.
Zurück zum Zitat Martino L, Masini M, Bugliani M, et al. Mast cells infiltrate pancreatic islets in human type 1 diabetes. Diabetologia. 2015;58:2554–62.PubMed Martino L, Masini M, Bugliani M, et al. Mast cells infiltrate pancreatic islets in human type 1 diabetes. Diabetologia. 2015;58:2554–62.PubMed
31.
Zurück zum Zitat Svensson J, Eising S, Mortensen HB, et al. High levels of immunoglobulin E and a continuous increase in immunoglobulin G and immunoglobulin M by age in children with newly diagnosed type 1 diabetes. Hum Immunol. 2012;73:17–25.PubMed Svensson J, Eising S, Mortensen HB, et al. High levels of immunoglobulin E and a continuous increase in immunoglobulin G and immunoglobulin M by age in children with newly diagnosed type 1 diabetes. Hum Immunol. 2012;73:17–25.PubMed
32.
Zurück zum Zitat Geoffrey R, Jia S, Kwitek AE, et al. Evidence of a functional role for mast cells in the development of type 1 diabetes mellitus in the BioBreeding rat. J Immunol. 2006;177:7275–86.PubMed Geoffrey R, Jia S, Kwitek AE, et al. Evidence of a functional role for mast cells in the development of type 1 diabetes mellitus in the BioBreeding rat. J Immunol. 2006;177:7275–86.PubMed
33.
Zurück zum Zitat Oka T, Kalesnikoff J, Starkl P, Tsai M, Galli SJ. Evidence questioning cromolyn’s effectiveness and selectivity as a ‘mast cell stabilizer’ in mice. Lab Investig J Tech Methods Pathol. 2012;92:1472–82. Oka T, Kalesnikoff J, Starkl P, Tsai M, Galli SJ. Evidence questioning cromolyn’s effectiveness and selectivity as a ‘mast cell stabilizer’ in mice. Lab Investig J Tech Methods Pathol. 2012;92:1472–82.
34.
Zurück zum Zitat Carlos D, Yaochite JN, Rocha FA, et al. Mast cells control insulitis and increase Treg cells to confer protection against STZ-induced type 1 diabetes in mice. Eur J Immunol. 2015;45:2873–85.PubMed Carlos D, Yaochite JN, Rocha FA, et al. Mast cells control insulitis and increase Treg cells to confer protection against STZ-induced type 1 diabetes in mice. Eur J Immunol. 2015;45:2873–85.PubMed
35.
Zurück zum Zitat Gutierrez DA, Fu W, Schonefeldt S, et al. Type 1 diabetes in NOD mice unaffected by mast cell deficiency. Diabetes. 2014;63:3827–34.PubMed Gutierrez DA, Fu W, Schonefeldt S, et al. Type 1 diabetes in NOD mice unaffected by mast cell deficiency. Diabetes. 2014;63:3827–34.PubMed
36.
Zurück zum Zitat Betto E, Usuelli V, Mandelli A, et al. Mast cells contribute to autoimmune diabetes by releasing interleukin-6 and failing to acquire a tolerogenic IL-10+ phenotype. Clin Immunol. 2017;178:29–38.PubMed Betto E, Usuelli V, Mandelli A, et al. Mast cells contribute to autoimmune diabetes by releasing interleukin-6 and failing to acquire a tolerogenic IL-10+ phenotype. Clin Immunol. 2017;178:29–38.PubMed
37.
Zurück zum Zitat Hübner MP, Larson D, Torrero MN, et al. Anti-FcεR1 antibody injections activate basophils and mast cells and delay type 1 diabetes onset in NOD mice. Clin Immunol. 2011;141:205–17.PubMedPubMedCentral Hübner MP, Larson D, Torrero MN, et al. Anti-FcεR1 antibody injections activate basophils and mast cells and delay type 1 diabetes onset in NOD mice. Clin Immunol. 2011;141:205–17.PubMedPubMedCentral
38.
Zurück zum Zitat Valle A, Giamporcaro GM, Scavini M, et al. Reduction of circulating neutrophils precedes and accompanies type 1 diabetes. Diabetes. 2013;62:2072–7.PubMedPubMedCentral Valle A, Giamporcaro GM, Scavini M, et al. Reduction of circulating neutrophils precedes and accompanies type 1 diabetes. Diabetes. 2013;62:2072–7.PubMedPubMedCentral
39.
Zurück zum Zitat Komi DEA, Shafaghat F, Christian M. Crosstalk between mast cells and adipocytes in physiologic and pathologic conditions. Clin Rev Allergy Immunol. 2020;2020:1–13. Komi DEA, Shafaghat F, Christian M. Crosstalk between mast cells and adipocytes in physiologic and pathologic conditions. Clin Rev Allergy Immunol. 2020;2020:1–13.
40.
Zurück zum Zitat Oishi Y, Manabe I. Integrated regulation of the cellular metabolism and function of immune cells in adipose tissue. Clin Exp Pharmacol Physiol. 2016;43:294–303.PubMed Oishi Y, Manabe I. Integrated regulation of the cellular metabolism and function of immune cells in adipose tissue. Clin Exp Pharmacol Physiol. 2016;43:294–303.PubMed
41.
Zurück zum Zitat Zatterale F, Longo M, Naderi J, et al. Chronic adipose tissue inflammation linking obesity to insulin resistance and type 2 diabetes. Front Physiol. 2020;10:1607.PubMedPubMedCentral Zatterale F, Longo M, Naderi J, et al. Chronic adipose tissue inflammation linking obesity to insulin resistance and type 2 diabetes. Front Physiol. 2020;10:1607.PubMedPubMedCentral
42.
Zurück zum Zitat Żelechowska P, Wiktorska M, Różalska S, et al. Leptin receptor is expressed by tissue mast cells. Immunol Res. 2018;66:557–66.PubMedPubMedCentral Żelechowska P, Wiktorska M, Różalska S, et al. Leptin receptor is expressed by tissue mast cells. Immunol Res. 2018;66:557–66.PubMedPubMedCentral
43.
Zurück zum Zitat Divoux A, Moutel S, Poitou C, et al. Mast cells in human adipose tissue: link with morbid obesity, inflammatory status, and diabetes. J Clin Endocrinol Metabol. 2012;97:E1677–85. Divoux A, Moutel S, Poitou C, et al. Mast cells in human adipose tissue: link with morbid obesity, inflammatory status, and diabetes. J Clin Endocrinol Metabol. 2012;97:E1677–85.
44.
Zurück zum Zitat Poglio S, De Toni-Costes F, Arnaud E, et al. Adipose tissue as a dedicated reservoir of functional mast cell progenitors. Stem Cells. 2010;28:2065–72.PubMed Poglio S, De Toni-Costes F, Arnaud E, et al. Adipose tissue as a dedicated reservoir of functional mast cell progenitors. Stem Cells. 2010;28:2065–72.PubMed
45.
Zurück zum Zitat Liu J, Divoux A, Sun J, et al. Genetic deficiency and pharmacological stabilization of mast cells reduce diet-induced obesity and diabetes in mice. Nat Med. 2009;15:940–5.PubMedPubMedCentral Liu J, Divoux A, Sun J, et al. Genetic deficiency and pharmacological stabilization of mast cells reduce diet-induced obesity and diabetes in mice. Nat Med. 2009;15:940–5.PubMedPubMedCentral
46.
Zurück zum Zitat Zhou Y, Yu X, Chen H, et al. Leptin deficiency shifts mast cells toward anti-inflammatory actions and protects mice from obesity and diabetes by polarizing M2 macrophages. Cell Metab. 2015;22:1045–58.PubMedPubMedCentral Zhou Y, Yu X, Chen H, et al. Leptin deficiency shifts mast cells toward anti-inflammatory actions and protects mice from obesity and diabetes by polarizing M2 macrophages. Cell Metab. 2015;22:1045–58.PubMedPubMedCentral
47.
Zurück zum Zitat Zhang X, Wang X, Yin H, et al. Functional inactivation of mast cells enhances subcutaneous adipose tissue browning in mice. Cell Rep. 2019;28(792–803):e794. Zhang X, Wang X, Yin H, et al. Functional inactivation of mast cells enhances subcutaneous adipose tissue browning in mice. Cell Rep. 2019;28(792–803):e794.
48.
Zurück zum Zitat Yabut JM, Desjardins EM, Chan EJ, et al. Genetic deletion of mast cell serotonin synthesis prevents the development of obesity and insulin resistance. Nature Commun. 2020;2020:11. Yabut JM, Desjardins EM, Chan EJ, et al. Genetic deletion of mast cell serotonin synthesis prevents the development of obesity and insulin resistance. Nature Commun. 2020;2020:11.
49.
Zurück zum Zitat Gutierrez DA, Muralidhar S, Feyerabend TB, Herzig S, Rodewald HR. Hematopoietic kit deficiency, rather than lack of mast cells, protects mice from obesity and insulin resistance. Cell Metab. 2015;21:678–91.PubMed Gutierrez DA, Muralidhar S, Feyerabend TB, Herzig S, Rodewald HR. Hematopoietic kit deficiency, rather than lack of mast cells, protects mice from obesity and insulin resistance. Cell Metab. 2015;21:678–91.PubMed
50.
Zurück zum Zitat Chmelař J, Chatzigeorgiou A, Chung K-J, et al. No role for mast cells in obesity-related metabolic dysregulation. Front Immunol. 2016;7:524.PubMedPubMedCentral Chmelař J, Chatzigeorgiou A, Chung K-J, et al. No role for mast cells in obesity-related metabolic dysregulation. Front Immunol. 2016;7:524.PubMedPubMedCentral
51.
Zurück zum Zitat Einwallner E, Kiefer FW, Di Caro G, et al. Mast cells are not associated with systemic insulin resistance. Eur J Clin Invest. 2016;46:911–9.PubMed Einwallner E, Kiefer FW, Di Caro G, et al. Mast cells are not associated with systemic insulin resistance. Eur J Clin Invest. 2016;46:911–9.PubMed
52.
Zurück zum Zitat Altintas MM, Nayer B, Walford EC, et al. Leptin deficiency-induced obesity affects the density of mast cells in abdominal fat depots and lymph nodes in mice. Lipids Health Dis. 2012;11:21.PubMedPubMedCentral Altintas MM, Nayer B, Walford EC, et al. Leptin deficiency-induced obesity affects the density of mast cells in abdominal fat depots and lymph nodes in mice. Lipids Health Dis. 2012;11:21.PubMedPubMedCentral
53.
Zurück zum Zitat Hickey FB, Martin F. Role of the immune system in diabetic kidney disease. Curr Diab Rep. 2018;18:20.PubMed Hickey FB, Martin F. Role of the immune system in diabetic kidney disease. Curr Diab Rep. 2018;18:20.PubMed
54.
Zurück zum Zitat Zhang J, Shi GP. Mast cells and metabolic syndrome. Biochim Biophys Acta. 2012;1822:14–20.PubMed Zhang J, Shi GP. Mast cells and metabolic syndrome. Biochim Biophys Acta. 2012;1822:14–20.PubMed
55.
Zurück zum Zitat Yin DD, Luo JH, Zhao ZY, Liao YJ, Li Y. Tranilast prevents renal interstitial fibrosis by blocking mast cell infiltration in a rat model of diabetic kidney disease. Mol Med Rep. 2018;17:7356–64.PubMed Yin DD, Luo JH, Zhao ZY, Liao YJ, Li Y. Tranilast prevents renal interstitial fibrosis by blocking mast cell infiltration in a rat model of diabetic kidney disease. Mol Med Rep. 2018;17:7356–64.PubMed
56.
Zurück zum Zitat de Morais RB, do Couto-Muniz VP, Costa EN, et al. Mast cell population in the development of diabetic nephropathy: effects of renin angiotensin system inhibition. Biomed Pharmacother. 2018;107:1115–8.PubMed de Morais RB, do Couto-Muniz VP, Costa EN, et al. Mast cell population in the development of diabetic nephropathy: effects of renin angiotensin system inhibition. Biomed Pharmacother. 2018;107:1115–8.PubMed
57.
Zurück zum Zitat Spinas E, Kritas S, Saggini A, et al. Role of mast cells in atherosclerosis: a classical inflammatory disease. London: SAGE; 2014. Spinas E, Kritas S, Saggini A, et al. Role of mast cells in atherosclerosis: a classical inflammatory disease. London: SAGE; 2014.
58.
Zurück zum Zitat Uemura K, Kondo H, Ishii Y, et al. Mast cells play an important role in the pathogenesis of hyperglycemia-induced atrial fibrillation. J Cardiovasc Electrophysiol. 2016;27:981–9.PubMed Uemura K, Kondo H, Ishii Y, et al. Mast cells play an important role in the pathogenesis of hyperglycemia-induced atrial fibrillation. J Cardiovasc Electrophysiol. 2016;27:981–9.PubMed
59.
Zurück zum Zitat Patella V, de Crescenzo G, Lamparter-Schummert B, De Rosa G, Adt M, Marone G. Increased cardiac mast cell density and mediator release in patients with dilated cardiomyopathy. Inflamm Res. 1997;46:31–2.PubMed Patella V, de Crescenzo G, Lamparter-Schummert B, De Rosa G, Adt M, Marone G. Increased cardiac mast cell density and mediator release in patients with dilated cardiomyopathy. Inflamm Res. 1997;46:31–2.PubMed
60.
Zurück zum Zitat Patella V, Marino I, Arbustini E, et al. Stem cell factor in mast cells and increased mast cell density in idiopathic and ischemic cardiomyopathy. Circulation. 1998;97:971–8.PubMed Patella V, Marino I, Arbustini E, et al. Stem cell factor in mast cells and increased mast cell density in idiopathic and ischemic cardiomyopathy. Circulation. 1998;97:971–8.PubMed
61.
Zurück zum Zitat Huang ZG, Jin Q, Fan M, et al. Myocardial remodeling in diabetic cardiomyopathy associated with cardiac mast cell activation. PLoS One. 2013;8:e60827.PubMedPubMedCentral Huang ZG, Jin Q, Fan M, et al. Myocardial remodeling in diabetic cardiomyopathy associated with cardiac mast cell activation. PLoS One. 2013;8:e60827.PubMedPubMedCentral
62.
Zurück zum Zitat He A, Fang W, Zhao K, et al. Mast cell-deficiency protects mice from streptozotocin-induced diabetic cardiomyopathy. Transl Res. 2019;208:1–14.PubMedPubMedCentral He A, Fang W, Zhao K, et al. Mast cell-deficiency protects mice from streptozotocin-induced diabetic cardiomyopathy. Transl Res. 2019;208:1–14.PubMedPubMedCentral
63.
Zurück zum Zitat Gurtner GC, Werner S, Barrandon Y, Longaker MT. Wound repair and regeneration. Nature. 2008;453:314–21.PubMed Gurtner GC, Werner S, Barrandon Y, Longaker MT. Wound repair and regeneration. Nature. 2008;453:314–21.PubMed
64.
Zurück zum Zitat Wallace HA, Basehore BM, Zito PM. Wound healing phases. In: StatPearls Treasure Island (FL). 2020. Wallace HA, Basehore BM, Zito PM. Wound healing phases. In: StatPearls Treasure Island (FL). 2020.
65.
Zurück zum Zitat Egozi EI, Ferreira AM, Burns AL, Gamelli RL, Dipietro LA. Mast cells modulate the inflammatory but not the proliferative response in healing wounds. Wound Repair Regenerat. 2003;11:46–54. Egozi EI, Ferreira AM, Burns AL, Gamelli RL, Dipietro LA. Mast cells modulate the inflammatory but not the proliferative response in healing wounds. Wound Repair Regenerat. 2003;11:46–54.
66.
Zurück zum Zitat Theoharides TC, Donelan JM, Papadopoulou N, Cao J, Kempuraj D, Conti P. Mast cells as targets of corticotropin-releasing factor and related peptides. Trends Pharmacol Sci. 2004;25:563–8.PubMed Theoharides TC, Donelan JM, Papadopoulou N, Cao J, Kempuraj D, Conti P. Mast cells as targets of corticotropin-releasing factor and related peptides. Trends Pharmacol Sci. 2004;25:563–8.PubMed
67.
Zurück zum Zitat Weller K, Foitzik K, Paus R, Syska W, Maurer M. Mast cells are required for normal healing of skin wounds in mice. FASEB J. 2006;20:2366–8.PubMed Weller K, Foitzik K, Paus R, Syska W, Maurer M. Mast cells are required for normal healing of skin wounds in mice. FASEB J. 2006;20:2366–8.PubMed
68.
Zurück zum Zitat Noli C, Miolo A. The mast cell in wound healing. Vet Dermatol. 2001;12:303–13.PubMed Noli C, Miolo A. The mast cell in wound healing. Vet Dermatol. 2001;12:303–13.PubMed
69.
Zurück zum Zitat Antsiferova M, Martin C, Huber M, et al. Mast cells are dispensable for normal and activin-promoted wound healing and skin carcinogenesis. J Immunol. 2013;191:6147–55.PubMed Antsiferova M, Martin C, Huber M, et al. Mast cells are dispensable for normal and activin-promoted wound healing and skin carcinogenesis. J Immunol. 2013;191:6147–55.PubMed
70.
Zurück zum Zitat Sehra S, Serezani AP, Ocaña JA, Travers JB, Kaplan MH. Mast cells regulate epidermal barrier function and the development of allergic skin inflammation. J Investig Dermatol. 2016;136:1429–37.PubMed Sehra S, Serezani AP, Ocaña JA, Travers JB, Kaplan MH. Mast cells regulate epidermal barrier function and the development of allergic skin inflammation. J Investig Dermatol. 2016;136:1429–37.PubMed
71.
Zurück zum Zitat Shubin NJ, Glukhova VA, Clauson M, et al. Proteome analysis of mast cell releasates reveals a role for chymase in the regulation of coagulation factor XIIIA levels via proteolytic degradation. J Allergy Clin Immunol. 2017;139:323–34.PubMed Shubin NJ, Glukhova VA, Clauson M, et al. Proteome analysis of mast cell releasates reveals a role for chymase in the regulation of coagulation factor XIIIA levels via proteolytic degradation. J Allergy Clin Immunol. 2017;139:323–34.PubMed
72.
Zurück zum Zitat Wulff BC, Parent AE, Meleski MA, DiPietro LA, Schrementi ME, Wilgus TA. Mast cells contribute to scar formation during fetal wound healing. J Invest Dermatol. 2012;132:458–65.PubMed Wulff BC, Parent AE, Meleski MA, DiPietro LA, Schrementi ME, Wilgus TA. Mast cells contribute to scar formation during fetal wound healing. J Invest Dermatol. 2012;132:458–65.PubMed
73.
Zurück zum Zitat Lamont P, Franklyn K, Rayman G, Boulton AJ. Update on the diabetic foot 2012: the 14th biennial Malvern Diabetic Foot Conference, May 9–11, 2012. Int J Lower Extremity Wounds. 2013;12:71–5. Lamont P, Franklyn K, Rayman G, Boulton AJ. Update on the diabetic foot 2012: the 14th biennial Malvern Diabetic Foot Conference, May 9–11, 2012. Int J Lower Extremity Wounds. 2013;12:71–5.
74.
Zurück zum Zitat Den Dekker A, Davis FM, Kunkel SL, Gallagher KA. Targeting epigenetic mechanisms in diabetic wound healing. Transl Res. 2019;204:39–50. Den Dekker A, Davis FM, Kunkel SL, Gallagher KA. Targeting epigenetic mechanisms in diabetic wound healing. Transl Res. 2019;204:39–50.
75.
Zurück zum Zitat Dinh T, Tecilazich F, Kafanas A, et al. Mechanisms involved in the development and healing of diabetic foot ulceration. Diabetes. 2012;61:2937–47.PubMedPubMedCentral Dinh T, Tecilazich F, Kafanas A, et al. Mechanisms involved in the development and healing of diabetic foot ulceration. Diabetes. 2012;61:2937–47.PubMedPubMedCentral
76.
Zurück zum Zitat Tellechea A, Kafanas A, Leal EC, et al. Increased skin inflammation and blood vessel density in human and experimental diabetes. Int J Low Extrem Wounds. 2013;12:4–11.PubMedPubMedCentral Tellechea A, Kafanas A, Leal EC, et al. Increased skin inflammation and blood vessel density in human and experimental diabetes. Int J Low Extrem Wounds. 2013;12:4–11.PubMedPubMedCentral
77.
Zurück zum Zitat Leal EC, Carvalho E, Tellechea A, et al. Substance P promotes wound healing in diabetes by modulating inflammation and macrophage phenotype. Am J Pathol. 2015;185:1638–48.PubMedPubMedCentral Leal EC, Carvalho E, Tellechea A, et al. Substance P promotes wound healing in diabetes by modulating inflammation and macrophage phenotype. Am J Pathol. 2015;185:1638–48.PubMedPubMedCentral
78.
Zurück zum Zitat Mantovani A, Sozzani S, Locati M, Allavena P, Sica A. Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol. 2002;23:549–55.PubMed Mantovani A, Sozzani S, Locati M, Allavena P, Sica A. Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol. 2002;23:549–55.PubMed
79.
80.
Zurück zum Zitat Sindrilaru A, Peters T, Wieschalka S, et al. An unrestrained proinflammatory M1 macrophage population induced by iron impairs wound healing in humans and mice. J Clin Investig. 2011;121:985–97.PubMed Sindrilaru A, Peters T, Wieschalka S, et al. An unrestrained proinflammatory M1 macrophage population induced by iron impairs wound healing in humans and mice. J Clin Investig. 2011;121:985–97.PubMed
81.
Zurück zum Zitat Novak ML, Koh TJ. Phenotypic transitions of macrophages orchestrate tissue repair. Am J Pathol. 2013;183:1352–63.PubMedPubMedCentral Novak ML, Koh TJ. Phenotypic transitions of macrophages orchestrate tissue repair. Am J Pathol. 2013;183:1352–63.PubMedPubMedCentral
82.
Zurück zum Zitat Theocharidis G, Bhasin S, Kounas K, Bhasin M, Veves A. Single cell RNA-seq analyses of healthy lower extremity skin and diabetic foot ulcers uncover distinct immune landscape of diabetic wound healing. Diabetes. 2018;67:647-P. Theocharidis G, Bhasin S, Kounas K, Bhasin M, Veves A. Single cell RNA-seq analyses of healthy lower extremity skin and diabetic foot ulcers uncover distinct immune landscape of diabetic wound healing. Diabetes. 2018;67:647-P.
83.
Zurück zum Zitat Tellechea A, Leal EC, Kafanas A, et al. Mast cells regulate wound healing in diabetes. Diabetes. 2016;65:2006–19.PubMedPubMedCentral Tellechea A, Leal EC, Kafanas A, et al. Mast cells regulate wound healing in diabetes. Diabetes. 2016;65:2006–19.PubMedPubMedCentral
84.
Zurück zum Zitat Strang H, Kaul A, Parikh U, et al. Chapter 11—role of cytokines and chemokines in wound healing. In: Bagchi D, Das A, Roy S, editors. Wound healing, tissue repair, and regeneration in diabetes. San Diego: Academic; 2020, p. 197–235. Strang H, Kaul A, Parikh U, et al. Chapter 11—role of cytokines and chemokines in wound healing. In: Bagchi D, Das A, Roy S, editors. Wound healing, tissue repair, and regeneration in diabetes. San Diego: Academic; 2020, p. 197–235.
85.
Zurück zum Zitat Bot I, Velden DV, Bouwman M, et al. Local mast cell activation promotes neovascularization. Cells. 2020;2020:9. Bot I, Velden DV, Bouwman M, et al. Local mast cell activation promotes neovascularization. Cells. 2020;2020:9.
86.
Zurück zum Zitat Nazari M, Ni NC, Lüdke A, et al. Mast cells promote proliferation and migration and inhibit differentiation of mesenchymal stem cells through PDGF. J Mol Cell Cardiol. 2016;94:32–42.PubMed Nazari M, Ni NC, Lüdke A, et al. Mast cells promote proliferation and migration and inhibit differentiation of mesenchymal stem cells through PDGF. J Mol Cell Cardiol. 2016;94:32–42.PubMed
87.
Zurück zum Zitat Dong X, Chen J, Zhang Y, Cen Y. Mast cell chymase promotes cell proliferation and expression of certain cytokines in a dose-dependent manner. Mol Med Rep. 2012;5:1487–90.PubMed Dong X, Chen J, Zhang Y, Cen Y. Mast cell chymase promotes cell proliferation and expression of certain cytokines in a dose-dependent manner. Mol Med Rep. 2012;5:1487–90.PubMed
88.
Zurück zum Zitat Vangansewinkel T, Geurts N, Quanten K, et al. Mast cells promote scar remodeling and functional recovery after spinal cord injury via mouse mast cell protease 6. FASEB J. 2016;30:2040–57.PubMed Vangansewinkel T, Geurts N, Quanten K, et al. Mast cells promote scar remodeling and functional recovery after spinal cord injury via mouse mast cell protease 6. FASEB J. 2016;30:2040–57.PubMed
89.
Zurück zum Zitat Gimenez-Rivera V-A, Siebenhaar F, Zimmermann C, Siiskonen H, Metz M, Maurer M. Mast cells limit the exacerbation of chronic allergic contact dermatitis in response to repeated allergen exposure. J Immunol. 2016;197:4240–6.PubMed Gimenez-Rivera V-A, Siebenhaar F, Zimmermann C, Siiskonen H, Metz M, Maurer M. Mast cells limit the exacerbation of chronic allergic contact dermatitis in response to repeated allergen exposure. J Immunol. 2016;197:4240–6.PubMed
90.
Zurück zum Zitat Balsam LB, DeAnda A Jr. The mast cell demystified: a novel target for anti-inflammatory strategies after circulatory arrest? J Thorac Cardiovasc Surg. 2017;153:77.PubMed Balsam LB, DeAnda A Jr. The mast cell demystified: a novel target for anti-inflammatory strategies after circulatory arrest? J Thorac Cardiovasc Surg. 2017;153:77.PubMed
91.
Zurück zum Zitat de Souza Junior DA, Santana C, Vieira GV, Oliver C, Jamur MC. Mast cell protease 7 promotes angiogenesis by degradation of integrin subunits. cells. 2019;8:349.PubMedCentral de Souza Junior DA, Santana C, Vieira GV, Oliver C, Jamur MC. Mast cell protease 7 promotes angiogenesis by degradation of integrin subunits. cells. 2019;8:349.PubMedCentral
92.
Zurück zum Zitat Perez-Favila A, Martinez-Fierro ML, Rodriguez-Lazalde JG, et al. Current therapeutic strategies in diabetic foot ulcers. Medicina. 2019;55:714.PubMedCentral Perez-Favila A, Martinez-Fierro ML, Rodriguez-Lazalde JG, et al. Current therapeutic strategies in diabetic foot ulcers. Medicina. 2019;55:714.PubMedCentral
93.
Zurück zum Zitat Gallant-Behm CL, Hildebrand KA, Hart DA. The mast cell stabilizer ketotifen prevents development of excessive skin wound contraction and fibrosis in red Duroc pigs. Wound Repair Regenerat. 2008;16:226–33. Gallant-Behm CL, Hildebrand KA, Hart DA. The mast cell stabilizer ketotifen prevents development of excessive skin wound contraction and fibrosis in red Duroc pigs. Wound Repair Regenerat. 2008;16:226–33.
94.
Zurück zum Zitat Chen L, Schrementi ME, Ranzer MJ, Wilgus TA, DiPietro LA. Blockade of mast cell activation reduces cutaneous scar formation. PLoS One. 2014;9:e85226.PubMedPubMedCentral Chen L, Schrementi ME, Ranzer MJ, Wilgus TA, DiPietro LA. Blockade of mast cell activation reduces cutaneous scar formation. PLoS One. 2014;9:e85226.PubMedPubMedCentral
95.
Zurück zum Zitat Tellechea A, Bai S, Dangwal S, et al. Topical application of a mast cell stabilizer improves impaired diabetic wound healing. J Invest Dermatol. 2020;140(901–911):e911. Tellechea A, Bai S, Dangwal S, et al. Topical application of a mast cell stabilizer improves impaired diabetic wound healing. J Invest Dermatol. 2020;140(901–911):e911.
96.
Zurück zum Zitat Kouhkheil R, Fridoni M, Abdollhifar M-A, et al. Impact of photobiomodulation and condition medium on mast cell counts, degranulation, and wound strength in infected skin wound healing of diabetic rats. Photobiomodul Photomed Laser Surg. 2019;37:706–14.PubMed Kouhkheil R, Fridoni M, Abdollhifar M-A, et al. Impact of photobiomodulation and condition medium on mast cell counts, degranulation, and wound strength in infected skin wound healing of diabetic rats. Photobiomodul Photomed Laser Surg. 2019;37:706–14.PubMed
97.
Zurück zum Zitat Babaei S, Ansarihadipour H, Nakhaei M, et al. Effect of Omegaven on mast cell concentration in diabetic wound healing. J Tissue Viabil. 2017;26:125–30. Babaei S, Ansarihadipour H, Nakhaei M, et al. Effect of Omegaven on mast cell concentration in diabetic wound healing. J Tissue Viabil. 2017;26:125–30.
98.
Zurück zum Zitat McLaughlin PJ, Cain JD, Titunick MB, Sassani JW, Zagon IS. Topical naltrexone is a safe and effective alternative to standard treatment of diabetic wounds. Adv Wound Care. 2017;6:279–88. McLaughlin PJ, Cain JD, Titunick MB, Sassani JW, Zagon IS. Topical naltrexone is a safe and effective alternative to standard treatment of diabetic wounds. Adv Wound Care. 2017;6:279–88.
99.
Zurück zum Zitat Orenstein S, Saberski E, Klueh U, Kreutzer D, Novitsky Y. Effects of mast cell modulation on early host response to implanted synthetic meshes. Hernia. 2010;14:511–6.PubMed Orenstein S, Saberski E, Klueh U, Kreutzer D, Novitsky Y. Effects of mast cell modulation on early host response to implanted synthetic meshes. Hernia. 2010;14:511–6.PubMed
Metadaten
Titel
Mast Cells in Diabetes and Diabetic Wound Healing
verfasst von
Jie Dong
Lihong Chen
Ying Zhang
Navin Jayaswal
Ikram Mezghani
Weijie Zhang
Aristidis Veves
Publikationsdatum
15.09.2020
Verlag
Springer Healthcare
Erschienen in
Advances in Therapy / Ausgabe 11/2020
Print ISSN: 0741-238X
Elektronische ISSN: 1865-8652
DOI
https://doi.org/10.1007/s12325-020-01499-4

Weitere Artikel der Ausgabe 11/2020

Advances in Therapy 11/2020 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Echinokokkose medikamentös behandeln oder operieren?

06.05.2024 DCK 2024 Kongressbericht

Die Therapie von Echinokokkosen sollte immer in spezialisierten Zentren erfolgen. Eine symptomlose Echinokokkose kann – egal ob von Hunde- oder Fuchsbandwurm ausgelöst – konservativ erfolgen. Wenn eine Op. nötig ist, kann es sinnvoll sein, vorher Zysten zu leeren und zu desinfizieren. 

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

Proximale Humerusfraktur: Auch 100-Jährige operieren?

01.05.2024 DCK 2024 Kongressbericht

Mit dem demographischen Wandel versorgt auch die Chirurgie immer mehr betagte Menschen. Von Entwicklungen wie Fast-Track können auch ältere Menschen profitieren und bei proximaler Humerusfraktur können selbst manche 100-Jährige noch sicher operiert werden.

Die „Zehn Gebote“ des Endokarditis-Managements

30.04.2024 Endokarditis Leitlinie kompakt

Worauf kommt es beim Management von Personen mit infektiöser Endokarditis an? Eine Kardiologin und ein Kardiologe fassen die zehn wichtigsten Punkte der neuen ESC-Leitlinie zusammen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.