[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
ijms-logo

Journal Browser

Journal Browser

Latest Review Papers in Molecular Immunology 2024

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Molecular Immunology".

Deadline for manuscript submissions: closed (20 December 2024) | Viewed by 19382

Special Issue Editors

Special Issue Information

Dear Colleagues,

This Special Issue aims to collect high-quality review papers discussing all the fields of molecular immunology. We encourage researchers from related fields to contribute review papers highlighting the latest developments in molecular immunology, or invite relevant experts and colleagues to do so. Full-length comprehensive reviews will be preferred.

Prof. Dr. Manlio Ferrarini
Prof. Dr. Vincenzo Barnaba
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • molecular immunology
  • review
  • immune cells

Benefits of Publishing in a Special Issue

  • Ease of navigation: Grouping papers by topic helps scholars navigate broad scope journals more efficiently.
  • Greater discoverability: Special Issues support the reach and impact of scientific research. Articles in Special Issues are more discoverable and cited more frequently.
  • Expansion of research network: Special Issues facilitate connections among authors, fostering scientific collaborations.
  • External promotion: Articles in Special Issues are often promoted through the journal's social media, increasing their visibility.
  • e-Book format: Special Issues with more than 10 articles can be published as dedicated e-books, ensuring wide and rapid dissemination.

Further information on MDPI's Special Issue polices can be found here.

Related Special Issues

Published Papers (10 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Review

35 pages, 2075 KiB  
Review
Inflammatory Effects and Regulatory Mechanisms of Chitinase-3-like-1 in Multiple Human Body Systems: A Comprehensive Review
by Dong Liu, Xin Hu, Xiao Ding, Ming Li and Lei Ding
Int. J. Mol. Sci. 2024, 25(24), 13437; https://doi.org/10.3390/ijms252413437 - 15 Dec 2024
Cited by 1 | Viewed by 575
Abstract
Chitinase-3-like-1 (Chi3l1), also known as YKL-40 or BRP-39, is a highly conserved mammalian chitinase with a chitin-binding ability but no chitinase enzymatic activity. Chi3l1 is secreted by various cell types and induced by several inflammatory cytokines. It can mediate a series of cell [...] Read more.
Chitinase-3-like-1 (Chi3l1), also known as YKL-40 or BRP-39, is a highly conserved mammalian chitinase with a chitin-binding ability but no chitinase enzymatic activity. Chi3l1 is secreted by various cell types and induced by several inflammatory cytokines. It can mediate a series of cell biological processes, such as proliferation, apoptosis, migration, differentiation, and polarization. Accumulating evidence has verified that Chi3l1 is involved in diverse inflammatory conditions; however, a systematic and comprehensive understanding of the roles and mechanisms of Chi3l1 in almost all human body system-related inflammatory diseases is still lacking. The human body consists of ten organ systems, which are combinations of multiple organs that perform one or more physiological functions. Abnormalities in these human systems can trigger a series of inflammatory environments, posing serious threats to the quality of life and lifespan of humans. Therefore, exploring novel and reliable biomarkers for these diseases is highly important, with Chi3l1 being one such parameter because of its physiological and pathophysiological roles in the development of multiple inflammatory diseases. Reportedly, Chi3l1 plays an important role in diagnosing and determining disease activity/severity/prognosis related to multiple human body system inflammation disorders. Additionally, many studies have revealed the influencing factors and regulatory mechanisms (e.g., the ERK and MAPK pathways) of Chi3l1 in these inflammatory conditions, identifying potential novel therapeutic targets for these diseases. In this review, we comprehensively summarize the potential roles and underlying mechanisms of Chi3l1 in inflammatory disorders of the respiratory, digestive, circulatory, nervous, urinary, endocrine, skeletal, muscular, and reproductive systems, which provides a more systematic understanding of Chi3l1 in multiple human body system-related inflammatory diseases. Moreover, this article summarizes potential therapeutic strategies for inflammatory diseases in these systems on the basis of the revealed roles and mechanisms mediated by Chi3l1. Full article
(This article belongs to the Special Issue Latest Review Papers in Molecular Immunology 2024)
Show Figures

Figure 1

Figure 1
<p>Summary of inflammatory diseases associated with elevated levels of Chi3l1 in various human body systems. Chi3l1 is highly expressed and acts as a valuable biomarker in inflammatory conditions affecting the nervous, digestive, endocrine, reproductive, muscular, circulatory, respiratory, urinary, and skeletal systems. The arrow indicates the increased Chi3l1 levels.</p>
Full article ">Figure 2
<p>Summary of the regulatory mechanisms by which Chi3l1 is involved in multiple human body system diseases. Various signaling pathways are involved in the regulation of Chi3l1 upregulation-related inflammatory diseases. The cascades associated with specific diseases of human organ systems (e.g., respiratory, digestive, circulatory, skeletal, nervous, endocrine, reproductive, and muscular systems) are shown in (<b>A</b>), and the detailed diagrams are shown in (<b>B</b>).</p>
Full article ">Figure 3
<p>Summary of therapeutic strategies involving Chi3l1 for multiple human body system diseases. Various therapeutic methods are used to alleviate inflammatory diseases of the respiratory, digestive, circulatory, skeletal, nervous, and endocrine systems by downregulating Chi3l1 levels.</p>
Full article ">
17 pages, 1012 KiB  
Review
Secreted Phospholipases A2: Drivers of Inflammation and Cancer
by Ivan Hidalgo, Maria Alba Sorolla, Anabel Sorolla, Antonieta Salud and Eva Parisi
Int. J. Mol. Sci. 2024, 25(22), 12408; https://doi.org/10.3390/ijms252212408 - 19 Nov 2024
Viewed by 1076
Abstract
Secreted phospholipase 2 (sPLA2) is the largest family of phospholipase A2 (PLA2) enzymes with 11 mammalian isoforms. Each sPLA2 exhibits different localizations and specific properties, being involved in a very wide spectrum of biological processes. The enzymatic activity of sPLA2 has been well [...] Read more.
Secreted phospholipase 2 (sPLA2) is the largest family of phospholipase A2 (PLA2) enzymes with 11 mammalian isoforms. Each sPLA2 exhibits different localizations and specific properties, being involved in a very wide spectrum of biological processes. The enzymatic activity of sPLA2 has been well described; however, recent findings have shown that they could regulate different signaling pathways by acting directly as ligands. Arachidonic acid (AA) and its derivatives are produced by sPLA2 in collaboration with other molecules in the extracellular space, making important impacts on the cellular environment, being especially relevant in the contexts of immunity and cancer. For these reasons, this review focuses on sPLA2 functions in processes such as the promotion of EMT, angiogenesis, and immunomodulation in the context of tumor initiation and progression. Finally, we will also describe how this knowledge has been applied in the search for new sPLA2 inhibitory compounds that can be used for cancer treatment. Full article
(This article belongs to the Special Issue Latest Review Papers in Molecular Immunology 2024)
Show Figures

Figure 1

Figure 1
<p>Metabolic products of arachidonic acid (AA). AA is released from membrane phospholipids by phospholipase A2 (PLA2) and metabolized by three main enzyme families: cyclooxygenases (COX), lipoxygenases (LOX), and cytochrome P450 (CYP450). COX-1 and COX-2 convert AA into prostaglandins (PG) D, I, E, A, and F2α, and thromboxanes A2 (TXA2). LOX catalyze the formation of hydroperoxyeicosatetraenoic acids (HpETE) and hydroxyeicosatetraenoic acids (HETE), leukotrienes (LT) A, B, C, D, and E, and lipoxins (LX) A and B. Lastly, CYP450 converts AA in epoxyeicosatrienoic acids (EET) and HETE. Dashed arrows represent each metabolite’s role in inflammation, angiogenesis, metastasis, proliferation, and immune modulation.</p>
Full article ">Figure 2
<p>Signaling networks that regulate epithelial–mesenchymal transition (EMT) activated by secreted phospholipases A2 (sPLA2). sPLA2 can activate EMT signaling pathways directly by engaging with sPLA2A-binding proteins (sPLA2A-BP) and being translocated to the cytoplasm, or by producing arachidonic acid and downstream metabolites (AA met). Transforming growth factor β (TGF-β), growth receptors and tumor necrosis factor α (TNFα) signaling pathways can induce EMT by the activation of the transcription factors (TF) SNAI1, ZEB1/2, TWIST, and SLUG. TGF-β induces EMT by the phosphorylation of Smad2 and Smad3, which localize to the nucleus with Smad4 to activate EMT TF. Several growth factors that act through tyrosine kinase receptors, such as epidermal growth factor (EGF), fibroblast growth factor (FGF), and hepatocyte growth factor (HGF), promote EMT thought the RAS-Mitogen-activated protein kinase (MAPK)/ERK signaling cascade or the Phosphatidylinositol 3-kinase/Protein kinase B (PI3K)/Akt axis, which ultimately activate nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). TNFα also activates the NF-κB pathway. Finally, Wnt stabilizes β-catenin, which translocates to the nucleus to activate ZEB1 and SNAI1 directly.</p>
Full article ">
27 pages, 1361 KiB  
Review
The Importance of Phosphoinositide 3-Kinase in Neuroinflammation
by Brock Wright, Samuel King and Cenk Suphioglu
Int. J. Mol. Sci. 2024, 25(21), 11638; https://doi.org/10.3390/ijms252111638 - 30 Oct 2024
Cited by 2 | Viewed by 1360
Abstract
Neuroinflammation, characterised by the activation of immune cells in the central nervous system (CNS), plays a dual role in both protecting against and contributing to the progression of neurodegenerative diseases, such as Alzheimer’s disease (AD) and multiple sclerosis (MS). This review explores the [...] Read more.
Neuroinflammation, characterised by the activation of immune cells in the central nervous system (CNS), plays a dual role in both protecting against and contributing to the progression of neurodegenerative diseases, such as Alzheimer’s disease (AD) and multiple sclerosis (MS). This review explores the role of phosphoinositide 3-kinase (PI3K), a key enzyme involved in cellular survival, proliferation, and inflammatory responses, within the context of neuroinflammation. Two PI3K isoforms of interest, PI3Kγ and PI3Kδ, are specific to the regulation of CNS cells, such as microglia, astrocytes, neurons, and oligodendrocytes, influencing pathways, such as Akt, mTOR, and NF-κB, that control cytokine production, immune cell activation, and neuroprotection. The dysregulation of PI3K signalling is implicated in chronic neuroinflammation, contributing to the exacerbation of neurodegenerative diseases. Preclinical studies show promise in targeting neuronal disorders using PI3K inhibitors, such as AS605240 (PI3Kγ) and idelalisib (PI3Kδ), which have reduced inflammation, microglial activation, and neuronal death in in vivo models of AD. However, the clinical translation of these inhibitors faces challenges, including blood–brain barrier (BBB) permeability, isoform specificity, and long-term safety concerns. This review highlights the therapeutic potential of PI3K modulation in neuroinflammatory diseases, identifying key gaps in the current research, particularly in the need for brain-penetrating and isoform-specific inhibitors. These findings underscore the importance of future research to develop targeted therapies that can effectively modulate PI3K activity and provide neuroprotection in chronic neurodegenerative disorders. Full article
(This article belongs to the Special Issue Latest Review Papers in Molecular Immunology 2024)
Show Figures

Figure 1

Figure 1
<p>PI3K signalling in neuroinflammation.</p>
Full article ">Figure 2
<p>Structural illustration of catalytic and regulatory subunits of PI3K Class I, II, and III. All PI3K catalytic subunits comprise a two-lobe (N and C) kinase, a helical domain, a lipid-binding C2 domain, and a Ras-binding domain (RBD). Class IA catalytic isoforms display an adaptor binding domain (ABD). Regulatory p85 group adaptors contain a SH3 domain at the N-terminal. Class IB PI3K isoforms possess adaptors of p101, sharing regions of low homology (H1 and H2). Class II PI3K exhibit extended N-terminal portions containing proline-rich motifs (P) followed by RBD, C2, and helical domains, along with Phox homology (PX) and an extra C2 domain. Class III contains C2 and helical domains; however, they lack RBD, instead binding to corresponding the p150 adaptor containing a kinase domain followed by heat (H) and aspartic/tryptofan40 (WD40) domains. Adapted from [<a href="#B35-ijms-25-11638" class="html-bibr">35</a>].</p>
Full article ">Figure 3
<p>The microglial PI3K pathway. RTK and GPCR activation from extracellular signals, growth factors, cytokines, and hormones, inducing tyrosine kinase domains aiding in the recruitment and activation of Class I PI3K. Phosphorylation of PI(4,5)P2 to PI(3,4,5)P3 leads to the activation of PDK1, Akt, and NF-<span class="html-italic">κ</span>B. The activation of NF-<span class="html-italic">κ</span>B induces gene expression of either pro- or anti-inflammatory mediators, potentially leading to an imbalance in proinflammatory mediators.</p>
Full article ">
17 pages, 1669 KiB  
Review
Post-Meningitic Syndrome: Pathophysiology and Consequences of Streptococcal Infections on the Central Nervous System
by Rachid Kaddoura, Karim Abdalbari, Mhmod Kadom, Beshr Abdulaziz Badla, Amin Abu Hijleh, Mohamed Hanifa, Masa AlAshkar, Mohamed Asbaita, Deema Othman, Hanan Faraji, Orjwan AlBakri, Sara Tahlak, Amir Abu Hijleh, Raneem Kabbani, Murtadha Resen, Helmi Abdalbari, Stefan S. Du Plessis and Temidayo S. Omolaoye
Int. J. Mol. Sci. 2024, 25(20), 11053; https://doi.org/10.3390/ijms252011053 - 15 Oct 2024
Viewed by 1410
Abstract
Streptococcus species represent a significant global cause of meningitis, leading to brain damage through bacterial virulence factors and the host inflammatory response. Upon entering the central nervous system (CNS), excessive inflammation leads to various neurological and psychological complications. This review explores the pathophysiological [...] Read more.
Streptococcus species represent a significant global cause of meningitis, leading to brain damage through bacterial virulence factors and the host inflammatory response. Upon entering the central nervous system (CNS), excessive inflammation leads to various neurological and psychological complications. This review explores the pathophysiological mechanisms and associated outcomes of streptococcal meningitis, particularly its short- and long-term neurological sequelae. Neurological symptoms, such as cognitive impairment, motor deficits, and sensory loss, are shown to vary in severity, with children being particularly susceptible to lasting complications. Among survivors, hearing loss, cognitive decline, and cranial nerve palsies emerge as the most frequently reported complications. The findings highlight the need for timely intervention, including neurorehabilitation strategies that focus on optimizing recovery and mitigating long-term disabilities. Future recommendations emphasize improving early diagnosis, expanding vaccine access, and personalizing rehabilitation protocols to enhance patient outcomes. As a novel contribution, this review proposes the term “post-meningitic syndrome” to showcase the broad spectrum of CNS complications that persist following streptococcal meningitis, providing a framework for a future clinical and research focus. Full article
(This article belongs to the Special Issue Latest Review Papers in Molecular Immunology 2024)
Show Figures

Figure 1

Figure 1
<p>Mechanism of CNS invasion by streptococcus species and BBB involvement. The figure illustrates the mechanism by which <span class="html-italic">Streptococcus pneumoniae</span> invades the BBB, causing damage that initially triggers a cytokine storm and increases oxidative stress. This sequence of events eventually leads to CNS damage, which can result in long-term CNS sequelae and potentially death. On the other side of the figure, the role of the immune system in combating the infection is depicted, showing how the bacteria activate the immune response, enabling host cells to eliminate the infection.</p>
Full article ">Figure 2
<p>Invasion and immune response to GAS and GBS.</p>
Full article ">Figure 3
<p>Multisystem complications of streptococcal infections.</p>
Full article ">
17 pages, 948 KiB  
Review
Molecular Mechanisms Affecting Statin Pharmacokinetics after Bariatric Surgery
by Matea Petrinović, Domagoj Majetić, Miro Bakula, Ivan Pećin, Daniela Fabris-Vitković, Marin Deškin, Deša Tešanović Perković, Maja Bakula, Marina Gradišer, Ines Bilić Ćurčić and Silvija Canecki-Varžić
Int. J. Mol. Sci. 2024, 25(19), 10375; https://doi.org/10.3390/ijms251910375 - 26 Sep 2024
Viewed by 1177
Abstract
According to recent data, one in eight people in the world struggle with obesity. Obesity management is increasingly dependent on bariatric surgical interventions, as the combination of lifestyle modifications and pharmacotherapy could have a modest long-term effect. Surgery is recommended only for individuals [...] Read more.
According to recent data, one in eight people in the world struggle with obesity. Obesity management is increasingly dependent on bariatric surgical interventions, as the combination of lifestyle modifications and pharmacotherapy could have a modest long-term effect. Surgery is recommended only for individuals whose body mass index (BMI) ≥ 40 kg/m2 and ≥ 35 kg/m2 in the presence of weight-related comorbidities. The most commonly performed procedures are sleeve gastrectomy and roux-en-Y gastric bypass. Pharmacokinetic and pharmacodynamic alterations occur as a result of the anatomical and physiological changes caused by surgery, which further differ depending on physicochemical drug factors and factors related to the dosage form. The following modifications are distinguished based on the type of bariatric surgery performed. Most bariatric patients have accompanying comorbidities, including dyslipidemia treated with hydroxymethylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors or statins. Significant improvements in the lipid profile are observed early in the postoperative period. The data reported in this review on statin pharmacokinetic alterations have demonstrated substantial inter- and intravariability, making it difficult to adopt clear guidelines. Based on the current literature review, reducing the statin dose to the lowest effective with continuous monitoring is considered an optimal approach in clinical practice. Full article
(This article belongs to the Special Issue Latest Review Papers in Molecular Immunology 2024)
Show Figures

Figure 1

Figure 1
<p>Molecular changes affecting drug pharmacokinetics after bariatric surgery.</p>
Full article ">Figure 2
<p>Summary of factors contributing to modified oral bioavailability of statin therapy following bariatric surgery. BS: bariatric surgery.</p>
Full article ">
16 pages, 1357 KiB  
Review
Inflammation, Autoimmunity, and Infection in Fibromyalgia: A Narrative Review
by Marino Paroli, Chiara Gioia, Daniele Accapezzato and Rosalba Caccavale
Int. J. Mol. Sci. 2024, 25(11), 5922; https://doi.org/10.3390/ijms25115922 - 29 May 2024
Cited by 4 | Viewed by 3237
Abstract
Fibromyalgia (FM) is a chronic disease characterized by widespread musculoskeletal pain of unknown etiology. The condition is commonly associated with other symptoms, including fatigue, sleep disturbances, cognitive impairment, and depression. For this reason, FM is also referred to as FM syndrome. The nature [...] Read more.
Fibromyalgia (FM) is a chronic disease characterized by widespread musculoskeletal pain of unknown etiology. The condition is commonly associated with other symptoms, including fatigue, sleep disturbances, cognitive impairment, and depression. For this reason, FM is also referred to as FM syndrome. The nature of the pain is defined as nociplastic according to the latest international classification and is characterized by altered nervous sensitization both centrally and peripherally. Psychosocial conditions have traditionally been considered critical in the genesis of FM. However, recent studies in animal models and humans have provided new evidence in favor of an inflammatory and/or autoimmune pathogenesis. In support of this hypothesis are epidemiological data of an increased female prevalence, similar to that of autoimmune diseases, and the frequent association with immune-mediated inflammatory disorders. In addition, the observation of an increased incidence of this condition during long COVID revived the hypothesis of an infectious pathogenesis. This narrative review will, therefore, discuss the evidence supporting the immune-mediated pathogenesis of FM in light of the most current data available in the literature. Full article
(This article belongs to the Special Issue Latest Review Papers in Molecular Immunology 2024)
Show Figures

Figure 1

Figure 1
<p>Passive transfer of serum IgG from fibromyalgia patients into mice induces fibromyalgia-like pain. Goebel et al. [<a href="#B122-ijms-25-05922" class="html-bibr">122</a>] recently demonstrated in an elegant experimental model that passive transfer of IgG-class immunoglobulin from fibromyalgia patients to mice induced sensory hypersensitivity through sensitization of nociceptive neurons. This experiment represented a breakthrough in understanding the pathogenesis of fibromyalgia, strongly suggesting an autoimmune mechanism mediated by antibodies against satellite glial cells and neurons.</p>
Full article ">Figure 2
<p>Induction of nociplastic pain by cells of the immune system. Cells of the innate and adaptive immune systems may participate in the genesis of nociplastic pain in FM. Microglia cells, neutrophils, and mast cells produce interleukins and chemokines. B cells produce autoantibodies against nerve cells. T cells produce both inflammatory cytokines, such as IFN-γ and IL-17, and anti-inflammatory cytokines, such as IL-10. NK cells are believed to inhibit pain through their stimulation of Mu-type opioid receptors. Viral and bacterial pathogens can act as stimulators of the immune system. The red circle banned sign above the arrow indicates the inhibitory action by the cell.</p>
Full article ">Figure 3
<p>Factors participating in the pathogenesis of fibromyalgia. Several factors are involved in the genesis of fibromyalgia (FM). Traditionally, psychosocial stress has been considered the main event in individuals predisposed to the activation of both central and peripheral pain sensitization. However, recent findings have demonstrated the key role played by the immune system. Inflammation mediated by mast cells, neutrophils, microglia cells, and natural killer (NK) cells produces several proinflammatory cytokines and chemokines that contribute to neuroinflammation and the subsequent increase in pain sensitization. On the other hand, recent studies have also involved adaptive immunity, demonstrating the role of T cells, particularly T helper (Th)-1 and Th17 cells capable of producing pro-inflammatory cytokines, and B cells through the production of neuron-specific autoantibodies, as demonstrated through animal models of passive IgG transfer in experimental animals. Finally, infections play an important role. In particular, infection with SARS-CoV-2, the causative agent of COVID-19, is believed to be responsible, through still unknown mechanisms, for the increased incidence of FM reported during the so-called “long COVID”. Once grafted, fibromyalgia has a chronic course and, in addition to widespread musculoskeletal pain, is accompanied by various debilitating symptoms such as depression, fatigue, cognitive impairment, also referred to as fibro fog, and sleep disturbances.</p>
Full article ">
14 pages, 1378 KiB  
Review
New Insight into Intestinal Mast Cells Revealed by Single-Cell RNA Sequencing
by Erisa Putro, Alessia Carnevale, Caterina Marangio, Valerio Fulci, Rossella Paolini and Rosa Molfetta
Int. J. Mol. Sci. 2024, 25(11), 5594; https://doi.org/10.3390/ijms25115594 - 21 May 2024
Viewed by 2045
Abstract
Mast cells (MCs) are tissue-resident immune cells distributed in all tissues and strategically located close to blood and lymphatic vessels and nerves. Thanks to the expression of a wide array of receptors, MCs act as tissue sentinels, able to detect the presence of [...] Read more.
Mast cells (MCs) are tissue-resident immune cells distributed in all tissues and strategically located close to blood and lymphatic vessels and nerves. Thanks to the expression of a wide array of receptors, MCs act as tissue sentinels, able to detect the presence of bacteria and parasites and to respond to different environmental stimuli. MCs originate from bone marrow (BM) progenitors that enter the circulation and mature in peripheral organs under the influence of microenvironment factors, thus differentiating into heterogeneous tissue-specific subsets. Even though MC activation has been traditionally linked to IgE-mediated allergic reactions, a role for these cells in other pathological conditions including tumor progression has recently emerged. However, several aspects of MC biology remain to be clarified. The advent of single-cell RNA sequencing platforms has provided the opportunity to understand MCs’ origin and differentiation as well as their phenotype and functions within different tissues, including the gut. This review recapitulates how single-cell transcriptomic studies provided insight into MC development as well as into the functional role of intestinal MC subsets in health and disease. Full article
(This article belongs to the Special Issue Latest Review Papers in Molecular Immunology 2024)
Show Figures

Figure 1

Figure 1
<p>MC origin and tissue heterogeneity analyzed by single-cell RNA sequencing (RNAseq). Single-cell RNAseq offers the possibility to identify the transcriptomic profiles of several cells from a tissue of interest. Transcripts associated with individual cells are sequenced and analyzed, resulting in cell clustering based on gene expression. (<b>A</b>) Identification of MC progenitors in bone marrow, fetal liver, and yolk sac has defined MC developmental trajectories; (<b>B</b>) gene expression profiles of MCs resident in different organs have clarified MC tissue heterogeneity; (<b>C</b>) transcript analysis of intestinal MCs has provided information on MC phenotypical and functional plasticity in health and disease. Created using BioRender.com.</p>
Full article ">Figure 2
<p>Intestinal MC phenotype and functions in homeostasis and inflammatory conditions. In healthy colons, two MC subsets have been identified: mucosal MC (MMC) and connective tissue-like MC (CTMC). In disease states, distinct MC subsets with unique gene expression profiles contribute to intestinal inflammation, as highlighted by different transcriptomic approaches. UC: ulcerative colitis; CD: celiac disease. Created using BioRender.com.</p>
Full article ">
18 pages, 2942 KiB  
Review
The Functional Roles of the Src Homology 2 Domain-Containing Inositol 5-Phosphatases SHIP1 and SHIP2 in the Pathogenesis of Human Diseases
by Spike Murphy Müller and Manfred Jücker
Int. J. Mol. Sci. 2024, 25(10), 5254; https://doi.org/10.3390/ijms25105254 - 11 May 2024
Cited by 1 | Viewed by 2155
Abstract
The src homology 2 domain-containing inositol 5-phosphatases SHIP1 and SHIP2 are two proteins involved in intracellular signaling pathways and have been linked to the pathogenesis of several diseases. Both protein paralogs are well known for their involvement in the formation of various kinds [...] Read more.
The src homology 2 domain-containing inositol 5-phosphatases SHIP1 and SHIP2 are two proteins involved in intracellular signaling pathways and have been linked to the pathogenesis of several diseases. Both protein paralogs are well known for their involvement in the formation of various kinds of cancer. SHIP1, which is expressed predominantly in hematopoietic cells, has been implicated as a tumor suppressor in leukemogenesis especially in myeloid leukemia, whereas SHIP2, which is expressed ubiquitously, has been implicated as an oncogene in a wider variety of cancer types and is suggested to be involved in the process of metastasis of carcinoma cells. However, there are numerous other diseases, such as inflammatory diseases as well as allergic responses, Alzheimer’s disease, and stroke, in which SHIP1 can play a role. Moreover, SHIP2 overexpression was shown to correlate with opsismodysplasia and Alzheimer’s disease, as well as metabolic diseases. The SHIP1-inhibitor 3-α-aminocholestane (3AC), and SHIP1-activators, such as AQX-435 and AQX-1125, and SHIP2-inhibitors, such as K161 and AS1949490, have been developed and partly tested in clinical trials, which indicates the importance of the SHIP-paralogs as possible targets in the therapy of those diseases. The aim of this article is to provide an overview of the current knowledge about the involvement of SHIP proteins in the pathogenesis of cancer and other human diseases and to create awareness that SHIP1 and SHIP2 are more than just tumor suppressors and oncogenes. Full article
(This article belongs to the Special Issue Latest Review Papers in Molecular Immunology 2024)
Show Figures

Figure 1

Figure 1
<p>AlphaFold protein structure predictions of the consensus sequences of SHIP1, with Va-lin117, and SHIP2 [<a href="#B7-ijms-25-05254" class="html-bibr">7</a>,<a href="#B8-ijms-25-05254" class="html-bibr">8</a>] (created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>). In this figure, the order of the domains from top to bottom is equivalent to the order of the domains within the corresponding protein from carboxy-terminus to amino-terminus.</p>
Full article ">Figure 2
<p>SHIP1 and SHIP2 in the PI3K/AKT signaling pathway (created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>).</p>
Full article ">Figure 3
<p>Involvement of SHIP1 in human diseases (created with BioRender). The numbers herein refer to the paragraphs with the corresponding subheading numbers under “3. The role of SHIP1 in human diseases”, wherein the involvement of SHIP1 in the pathogenesis of those diseases is further elucidated.</p>
Full article ">Figure 4
<p>Pharmacological SHIP paralog regulators (created with BioRender). API regulates the function of SHIP1 via inhibition of miR-155. AQX-1125 has been tested in clinical studies.</p>
Full article ">Figure 5
<p>Involvement of SHIP2 in human diseases (created with BioRender). The numbers herein refer to the paragraphs with the corresponding subheading numbers under “5. The role of SHIP2 in human diseases”, wherein the involvement of SHIP2 in the pathogenesis of those diseases is further elucidated.</p>
Full article ">
15 pages, 1102 KiB  
Review
The Golgi Apparatus: A Key Player in Innate Immunity
by Ion Mărunţelu, Alexandra-Elena Constantinescu, Razvan-Adrian Covache-Busuioc and Ileana Constantinescu
Int. J. Mol. Sci. 2024, 25(7), 4120; https://doi.org/10.3390/ijms25074120 - 8 Apr 2024
Cited by 6 | Viewed by 3094
Abstract
The Golgi apparatus, long recognized for its roles in protein processing and vesicular trafficking, has recently been identified as a crucial contributor to innate immune signaling pathways. This review discusses our expanding understanding of the Golgi apparatus’s involvement in initiating and activating these [...] Read more.
The Golgi apparatus, long recognized for its roles in protein processing and vesicular trafficking, has recently been identified as a crucial contributor to innate immune signaling pathways. This review discusses our expanding understanding of the Golgi apparatus’s involvement in initiating and activating these pathways. It highlights the significance of membrane connections between the Golgi and other organelles, such as the endoplasmic reticulum, mitochondria, endosomes, and autophagosomes. These connections are vital for the efficient transmission of innate immune signals and the activation of effector responses. Furthermore, the article delves into the Golgi apparatus’s roles in key immune pathways, including the inflammasome-mediated activation of caspase-1, the cGAS-STING pathway, and TLR/RLR signaling. Overall, this review aims to provide insights into the multifunctional nature of the Golgi apparatus and its impact on innate immunity. Full article
(This article belongs to the Special Issue Latest Review Papers in Molecular Immunology 2024)
Show Figures

Figure 1

Figure 1
<p>The figure shows PLC releases Ca<sup>2+</sup> from the ER through inositol-1, 4, 5-trisphosphate (InsP3) Receptor. Ca<sup>2+</sup> accumulates in the mitochondria, which may lead to mitochondrial damage (1). Damaged mitochondria release calcium ions (Ca<sup>2+</sup>) that accumulate in the mitochondria-associated membranes (MAMs), contributing to their activation (1*). Damaged mitochondria release several factors that trigger the activation of the NLRP3 inflammasome (2). NLRP3 was shown to directly bind to mitochondria-associated ER membranes (MAMs) (black arrow). The other product of PLC activation, diacylglycerol (DAG), accumulates in the Golgi (3) and increases the activity of PKD (protein kinase D), which then phosphorylates NLRP3-Inflammasome and activates it (3*), releasing it from MAMs (4).</p>
Full article ">Figure 2
<p>STING activates TBK1 at the Golgi. TBK1 phosphorylates interferon regulatory factor 3 (IRF3), leading to transcription of IFNb genes and stimulation of type I and III interferon genes as well as genes encoding proinflammatory cytokines and chemokines. STING is then degraded by the lysosomes through microautophagy.</p>
Full article ">
23 pages, 370 KiB  
Review
Role of CARD9 in Cell- and Organ-Specific Immune Responses in Various Infections
by Ji Seok Lee and Chaekyun Kim
Int. J. Mol. Sci. 2024, 25(5), 2598; https://doi.org/10.3390/ijms25052598 - 23 Feb 2024
Cited by 1 | Viewed by 1636
Abstract
The caspase recruitment domain-containing protein 9 (CARD9) is an intracellular adaptor protein that is abundantly expressed in cells of the myeloid lineage, such as neutrophils, macrophages, and dendritic cells. CARD9 plays a critical role in host immunity against infections caused by fungi, bacteria, [...] Read more.
The caspase recruitment domain-containing protein 9 (CARD9) is an intracellular adaptor protein that is abundantly expressed in cells of the myeloid lineage, such as neutrophils, macrophages, and dendritic cells. CARD9 plays a critical role in host immunity against infections caused by fungi, bacteria, and viruses. A CARD9 deficiency impairs the production of inflammatory cytokines and chemokines as well as migration and infiltration, thereby increasing susceptibility to infections. However, CARD9 signaling varies depending on the pathogen causing the infection. Furthermore, different studies have reported altered CARD9-mediated signaling even with the same pathogen. Therefore, this review focuses on and elucidates the current literature on varied CARD9 signaling in response to various infectious stimuli in humans and experimental mice models. Full article
(This article belongs to the Special Issue Latest Review Papers in Molecular Immunology 2024)
Back to TopTop