Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.376
Filter
1.
Front Immunol ; 15: 1334829, 2024.
Article in English | MEDLINE | ID: mdl-38827746

ABSTRACT

Infectious diseases continue to pose significant global health challenges. In addition to the enduring burdens of ailments like malaria and HIV, the emergence of nosocomial outbreaks driven by antibiotic-resistant pathogens underscores the ongoing threats. Furthermore, recent infectious disease crises, exemplified by the Ebola and SARS-CoV-2 outbreaks, have intensified the pursuit of more effective and efficient diagnostic and therapeutic solutions. Among the promising options, antibodies have garnered significant attention due to their favorable structural characteristics and versatile applications. Notably, nanobodies (Nbs), the smallest functional single-domain antibodies of heavy-chain only antibodies produced by camelids, exhibit remarkable capabilities in stable antigen binding. They offer unique advantages such as ease of expression and modification and enhanced stability, as well as improved hydrophilicity compared to conventional antibody fragments (antigen-binding fragments (Fab) or single-chain variable fragments (scFv)) that can aggregate due to their low solubility. Nanobodies directly target antigen epitopes or can be engineered into multivalent Nbs and Nb-fusion proteins, expanding their therapeutic potential. This review is dedicated to charting the progress in Nb research, particularly those derived from camelids, and highlighting their diverse applications in treating infectious diseases, spanning both human and animal contexts.


Subject(s)
Camelidae , Single-Domain Antibodies , Animals , Single-Domain Antibodies/immunology , Single-Domain Antibodies/therapeutic use , Humans , Camelidae/immunology , Communicable Diseases/immunology , Communicable Diseases/therapy , Camelids, New World/immunology , COVID-19/immunology , COVID-19/therapy
4.
Front Cell Infect Microbiol ; 14: 1384420, 2024.
Article in English | MEDLINE | ID: mdl-38756232

ABSTRACT

Infectious diseases are among the factors that account for a significant proportion of disease-related deaths worldwide. The primary treatment approach to combat microbial infections is the use of antibiotics. However, the widespread use of these drugs over the past two decades has led to the emergence of resistant microbial species, making the control of microbial infections a serious challenge. One of the most important solutions in the field of combating infectious diseases is the regulation of the host's defense system. Toll-like receptors (TLRs) play a crucial role in the first primary defense against pathogens by identifying harmful endogenous molecules released from dying cells and damaged tissues as well as invading microbial agents. Therefore, they play an important role in communicating and regulating innate and adaptive immunity. Of course, excessive activation of TLRs can lead to disruption of immune homeostasis and increase the risk of inflammatory reactions. Targeting TLR signaling pathways has emerged as a new therapeutic approach for infectious diseases based on host-directed therapy (HDT). In recent years, stem cell-derived exosomes have received significant attention as factors regulating the immune system. The regulation effects of exosomes on the immune system are based on the HDT strategy, which is due to their cargoes. In general, the mechanism of action of stem cell-derived exosomes in HDT is by regulating and modulating immunity, promoting tissue regeneration, and reducing host toxicity. One of their most important cargoes is microRNAs, which have been shown to play a significant role in regulating immunity through TLRs. This review investigates the therapeutic properties of stem cell-derived exosomes in combating infections through the interaction between exosomal microRNAs and Toll-like receptors.


Subject(s)
Exosomes , MicroRNAs , Stem Cells , Toll-Like Receptors , Exosomes/metabolism , Toll-Like Receptors/metabolism , Humans , MicroRNAs/metabolism , MicroRNAs/genetics , Animals , Stem Cells/metabolism , Signal Transduction , Immunity, Innate , Communicable Diseases/immunology , Communicable Diseases/metabolism , Adaptive Immunity
5.
World J Microbiol Biotechnol ; 40(7): 209, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38771414

ABSTRACT

Nanobodies are the smallest known antigen-binding molecules to date. Their small size, good tissue penetration, high stability and solubility, ease of expression, refolding ability, and negligible immunogenicity in the human body have granted them excellence over conventional antibodies. Those exceptional attributes of nanobodies make them promising candidates for various applications in biotechnology, medicine, protein engineering, structural biology, food, and agriculture. This review presents an overview of their structure, development methods, advantages, possible challenges, and applications with special emphasis on infectious diseases-related ones. A showcase of how nanobodies can be harnessed for applications including neutralization of viruses and combating antibiotic-resistant bacteria is detailed. Overall, the impact of nanobodies in vaccine design, rapid diagnostics, and targeted therapies, besides exploring their role in deciphering microbial structures and virulence mechanisms are highlighted. Indeed, nanobodies are reshaping the future of infectious disease prevention and treatment.


Subject(s)
Communicable Diseases , Single-Domain Antibodies , Single-Domain Antibodies/immunology , Humans , Communicable Diseases/immunology , Communicable Diseases/therapy , Animals , Biotechnology/methods , Protein Engineering/methods
6.
Life Sci ; 348: 122686, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38710282

ABSTRACT

Proper and functional immune response requires a complex interaction between innate and adaptive immune cells, which dendritic cells (DCs) are the primary actors in this coordination as professional antigen-presenting cells. DCs are armed with numerous pattern recognition receptors (PRRs) such as nucleotide-binding and oligomerization domain-like receptors (NLRs) like NLRP3, which influence the development of their activation state upon sensation of ligands. NLRP3 is a crucial component of the immune system for protection against tumors and infectious agents, because its activation leads to the assembly of inflammasomes that cause the formation of active caspase-1 and stimulate the maturation and release of proinflammatory cytokines. But, when NLRP3 becomes overactivated, it plays a pathogenic role in the progression of several autoimmune disorders. So, NLRP3 activation is strictly regulated by diverse signaling pathways that are mentioned in detail in this review. Furthermore, the role of NLRP3 in all of the diverse immune cells' subsets is briefly mentioned in this study because NLRP3 plays a pivotal role in modulating other immune cells which are accompanied by DCs' responses and subsequently influence differentiation of T cells to diverse T helper subsets and even impact on cytotoxic CD8+ T cells' responses. This review sheds light on the functional and therapeutic role of NLRP3 in DCs and its contribution to the occurrence and progression of autoimmune disorders, prevention of diverse tumors' development, and recognition and annihilation of various infectious agents. Furthermore, we highlight NLRP3 targeting potential for improving DC-based immunotherapeutic approaches, to be used for the benefit of patients suffering from these disorders.


Subject(s)
Autoimmune Diseases , Autoimmunity , Dendritic Cells , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Neoplasms , Dendritic Cells/immunology , Dendritic Cells/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Humans , Neoplasms/immunology , Neoplasms/therapy , Inflammasomes/immunology , Inflammasomes/metabolism , Animals , Autoimmunity/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Autoimmune Diseases/metabolism , Communicable Diseases/immunology , Communicable Diseases/metabolism , Communicable Diseases/therapy
7.
J Math Biol ; 88(6): 71, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38668894

ABSTRACT

In epidemics, waning immunity is common after infection or vaccination of individuals. Immunity levels are highly heterogeneous and dynamic. This work presents an immuno-epidemiological model that captures the fundamental dynamic features of immunity acquisition and wane after infection or vaccination and analyzes mathematically its dynamical properties. The model consists of a system of first order partial differential equations, involving nonlinear integral terms and different transfer velocities. Structurally, the equation may be interpreted as a Fokker-Planck equation for a piecewise deterministic process. However, unlike the usual models, our equation involves nonlocal effects, representing the infectivity of the whole environment. This, together with the presence of different transfer velocities, makes the proved existence of a solution novel and nontrivial. In addition, the asymptotic behavior of the model is analyzed based on the obtained qualitative properties of the solution. An optimal control problem with objective function including the total number of deaths and costs of vaccination is explored. Numerical results describe the dynamic relationship between contact rates and optimal solutions. The approach can contribute to the understanding of the dynamics of immune responses at population level and may guide public health policies.


Subject(s)
Communicable Diseases , Mathematical Concepts , Models, Immunological , Vaccination , Humans , Vaccination/statistics & numerical data , Communicable Diseases/immunology , Communicable Diseases/epidemiology , Communicable Diseases/transmission , Computer Simulation , Epidemics/statistics & numerical data , Epidemiological Models
8.
Science ; 384(6693): eadl2016, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38635718

ABSTRACT

Infectious diseases continue to claim many lives. Prevention of morbidity and mortality from these diseases would benefit not just from new medicines and vaccines but also from a better understanding of what constitutes protective immunity. Among the major immune signals that mobilize host defense against infection is interferon-γ (IFN-γ), a protein secreted by lymphocytes. Forty years ago, IFN-γ was identified as a macrophage-activating factor, and, in recent years, there has been a resurgent interest in IFN-γ biology and its role in human defense. Here we assess the current understanding of IFN-γ, revisit its designation as an "interferon," and weigh its prospects as a therapeutic against globally pervasive microbial pathogens.


Subject(s)
Communicable Diseases , Interferon-gamma , Humans , Communicable Diseases/immunology , Communicable Diseases/therapy , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interferon-gamma/therapeutic use , Immunity/genetics
9.
Steroids ; 207: 109426, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38685461

ABSTRACT

Multiple epidemiologic studies have revealed that gender is considered one of the important factors in the frequency and severity of certain infectious diseases, in which estrogens may play a vital role. There is growing evidence that estrogens as female sex hormone can modulate multiple biological functions outside of the reproductive system, such as in brain and cardiovascular system. However, it is largely unknown about the roles and mechanisms of estrogens/estrogen receptors in immune health and infection disease. Thence, by reading a lot of literature, we summarized the regulatory mechanisms of estrogens/estrogen receptors in immune cells and their roles in certain infectious diseases with gender differences. Therefore, estrogens may have therapeutic potentials to prevent and treat these infectious diseases, which needs further clinical investigation.


Subject(s)
Estrogens , Humans , Estrogens/metabolism , Estrogens/immunology , Animals , Receptors, Estrogen/metabolism , Immune System/metabolism , Immune System/immunology , Communicable Diseases/immunology , Communicable Diseases/metabolism , Infections/immunology
13.
Eur J Intern Med ; 123: 15-22, 2024 May.
Article in English | MEDLINE | ID: mdl-38135583

ABSTRACT

Upon encountering pathogens, the immune system typically responds by initiating an acute and self-limiting reaction, with symptoms subsiding after the pathogen has been cleared. However, long-term post-infectious clinical symptoms can manifest months or even years after the initial infection. 'Trained immunity', the functional reprogramming of innate immune cells through epigenetic and metabolic rewiring, has been proposed as a key concept for understanding these long-term effects. Although trained immunity can result in enhanced protection against reinfection with heterologous pathogens, it can also contribute to detrimental outcomes. Persisting and excessive inflammation can cause tissue damage and aggravate immune-mediated conditions and cardiovascular complications. On the other hand, suppression of immune cell effector functions by long-lasting epigenetic changes can result in post-infectious immune paralysis. Distinct stimuli can evoke different trained immunity programs, potentially resulting in different consequences for the host. In this review, we provide an overview of both the adaptive and maladaptive consequences of infectious diseases. We discuss how long-term immune dysregulation in patients can be addressed by tailoring host-directed interventions and identify areas of scientific and therapeutic potential to advance further.


Subject(s)
Immunity, Innate , Humans , Epigenesis, Genetic , Inflammation/immunology , Communicable Diseases/immunology , Adaptive Immunity
14.
Inmunología (1987) ; 42(2): 38-43, Jun.-Dic. 2023. ilus
Article in Spanish | IBECS | ID: ibc-231264

ABSTRACT

En este trabajo, recientemente publicado en la prestigiosa revista Advanced Science (Adv Sci; Weinheim, Baden-Württemberg, Germany), se ha diseñado y estudiado a nivel preclínico un anticuerpo biespecífico neutralizante (nAbs) frente al virus SARS-CoV-2 y dirigido específicamente a un receptor de células dendríticas convencionales de tipo 1 (cDC1s) para estimular específicamente la respuesta de linfocitos T citotóxicos. El trabajo surge de la colaboración de un consorcio muy amplio de grupos de investigación expertos en distintas áreas de la inmunología, biología celular, virología, biología estructural, química de proteínas y partículas, que han logrado generar un anticuerpo trimérico biespecífico, TNTDNGR-1. TNT se corresponde con las siglas en inglés para anticuerpo neutralizante en tándem trimérico, mientras que DNGR-1 es la molécula inmuno-reguladora “dendritic cell natural killer group receptor-1”, también conocida como CLEC9A. TNTDNGR-1 presenta una gran avidez frente a la región RBD (“receptor binding domain”) del virus SARS-CoV-2 y también frente al receptor DNGR-1, una lectina tipo C expresada por cDC1s. Se trata, además de una colaboración público-privada, con la participación de una compañía de biotecnología española, otorgando valor añadido al trabajo de investigación, por su potencial traslación a la clínica a corto o medio plazo. Mediante el uso de técnicas de crio-microscopía electrónica se ha comprobado que la estructura TNT , backbone de TNTDNGR-1, permite la unión simultánea a sus seis epítopos en la proteína S (del inglés spike) del SARS-CoV-2, dos por cada RBD, dotándola de una interacción neutralizante de alta afinidad frente al virus. ... (AU)


Subject(s)
Humans , Antibodies, Bispecific/immunology , /immunology , T-Lymphocytes, Cytotoxic/immunology , Dendritic Cells/immunology , Communicable Diseases/immunology
15.
Yi Chuan ; 45(11): 1007-1017, 2023 Nov 20.
Article in English | MEDLINE | ID: mdl-38764266

ABSTRACT

Guanylate-binding proteins (GBPs) are a subfamily of interferon-inducible proteins that undertake distinct roles in the the context of bacteria, virus, chlamydia and parasites infections. These proteins exert a notable influence on the progression and outcomes of infectious diseases. Within the realm of host cell-autonomous immunity against pathogens, GBPs have been identified as the regulators of pyroptosis through canonical and noncanonical inflammasome activation pathways. In this review, we summarize the structure and evolution of GBP family members, the canonical and noncanonical inflammasome activation pathways, the roles of GBPs in regulating inflammasome activation, and the mechanisms of GBPs affecting infections induced by different pathogens. We hope to provide new basic research clues for the pathogenesis and diagnosis and treatment of infectious diseases.


Subject(s)
GTP-Binding Proteins , Inflammasomes , Inflammasomes/immunology , Humans , Animals , GTP-Binding Proteins/genetics , GTP-Binding Proteins/immunology , Communicable Diseases/immunology , Communicable Diseases/genetics
16.
Int J Mol Sci ; 23(22)2022 Nov 17.
Article in English | MEDLINE | ID: mdl-36430676

ABSTRACT

Research of the last decade has remarkably increased our understanding of innate lymphoid cells (ILCs). ILCs, in analogy to T helper (Th) cells and their cytokine and transcription factor profile, are categorized into three distinct populations: ILC1s express the transcription factor T-bet and secrete IFNγ, ILC2s depend on the expression of GATA-3 and release IL-5 and IL-13, and ILC3s express RORγt and secrete IL-17 and IL-22. Noteworthy, ILCs maintain a level of plasticity, depending on exposed cytokines and environmental stimuli. Furthermore, ILCs are tissue resident cells primarily localized at common entry points for pathogens such as the gut-associated lymphoid tissue (GALT). They have the unique capacity to initiate rapid responses against pathogens, provoked by changes of the cytokine profile of the respective tissue. Moreover, they regulate tissue inflammation and homeostasis. In case of intracellular pathogens entering the mucosal tissue, ILC1s respond by secreting cytokines (e.g., IFNγ) to limit the pathogen spread. Upon infection with helminths, intestinal epithelial cells produce alarmins (e.g., IL-25) and activate ILC2s to secrete IL-13, which induces differentiation of intestinal stem cells into tuft and goblet cells, important for parasite expulsion. Additionally, during bacterial infection ILC3-derived IL-22 is required for bacterial clearance by regulating antimicrobial gene expression in epithelial cells. Thus, ILCs can limit infectious diseases via secretion of inflammatory mediators and interaction with other cell types. In this review, we will address the role of ILCs during enteric infectious diseases.


Subject(s)
Gastrointestinal Diseases , Immunity, Innate , Lymphocytes , Humans , Communicable Diseases/immunology , Communicable Diseases/metabolism , Cytokines/metabolism , Immunity, Innate/physiology , Interleukin-13/metabolism , Lymphocytes/metabolism , Gastrointestinal Diseases/immunology , Gastrointestinal Diseases/microbiology
17.
Proc Natl Acad Sci U S A ; 119(14): e2119093119, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35312341

ABSTRACT

SignificanceUsing SARS-CoV-2 as a relevant case study for infectious disease, we investigate the structure-function relationships that dictate antiviral spherical nucleic acid (SNA) vaccine efficacy. We show that the SNA architecture can be rapidly employed to target COVID-19 through incorporation of the receptor-binding domain, and that the resulting vaccine potently activates human cells in vitro and mice in vivo. Furthermore, when challenged with a lethal viral infection, only mice treated with the SNA vaccine survived. Taken together, this work underscores the importance of rational vaccine design for infectious disease to yield vaccines that elicit more potent immune responses to effectively fight disease.


Subject(s)
Communicable Disease Control , Nucleic Acids/immunology , Vaccines, DNA/immunology , Animals , Biotechnology , COVID-19/prevention & control , Communicable Disease Control/methods , Communicable Diseases/etiology , Communicable Diseases/immunology , Humans , Nucleic Acids/chemistry , SARS-CoV-2/immunology , Vaccine Development , Vaccines, DNA/genetics , Viral Vaccines/genetics , Viral Vaccines/immunology
18.
J Virol ; 96(7): e0009822, 2022 04 13.
Article in English | MEDLINE | ID: mdl-35285684

ABSTRACT

Respiratory viruses cause mild to severe diseases in humans every year, constituting a major public health problem. Characterizing the pathogenesis in physiologically relevant models is crucial for developing efficient vaccines and therapeutics. Here, we show that lung organoids derived from human primary or lung tumor tissue maintain the cellular composition and characteristics of the original tissue. Moreover, we show that these organoids sustain viral replication with particular infection foci formation, and they activate the expression of interferon-associated and proinflammatory genes responsible for mediating a robust innate immune response. All together, we show that three-dimensional (3D) lung organoids constitute a relevant platform to model diseases and enable the development of drug screenings. IMPORTANCE Three-dimensional (3D) human lung organoids reflect the native cell composition of the lung as well as its physiological properties. Human 3D lung organoids offer ideal conditions, such as timely availability in large quantities and high physiological relevance for reassessment and prediction of disease outbreaks of respiratory pathogens and pathogens that use the lung as a primary entry portal. Human lung organoids can be used in basic research and diagnostic settings as early warning cell culture systems and also serve as a relevant platform for modeling infectious diseases and drug development. They can be used to characterize pathogens and analyze the influence of infection on, for example, immunological parameters, such as the expression of interferon-associated and proinflammatory genes in the context of cancer. In our study, we found that cancer-derived lung organoids were more sensitive to influenza A virus infection than those derived from healthy tissue and demonstrated a decreased innate immune response.


Subject(s)
Lung , Organ Culture Techniques , Organoids , Communicable Diseases/diagnosis , Communicable Diseases/immunology , Humans , Immunity, Innate , Interferons , Lung/pathology , Organ Culture Techniques/methods , Organoids/immunology , Organoids/virology
19.
Carbohydr Res ; 513: 108527, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35240551

ABSTRACT

iNKT cells are a special type of T cell that acts as a link between the innate and adaptive immune systems, with the capacity to stimulate a wide range of cell types. The glycolipid α-galactosylceramide (αGC) is a robust agonist of iNKT cells and induces the secretion of Th1- and Th2-type cytokines. αGC and its analogs are widely used as adjuvants to enhance immune responses against viral, parasitic, and bacterial pathogens. This review first discusses the challenges of using free αGC as a vaccine adjuvant to treat infectious diseases. We next present strategies to realize the potential of the adjuvant effect of iNKT cell glycolipids, including (1) the use of Th1- or Th2-biasing αGC analogs, (2) covalent conjugation of glycolipid with antigen, (3) particulate vehicle-assisted delivery of glycolipid, (4) glycolipid-loaded cellular systems, (5) glycolipid combination with other immunostimulants, and (6) usage as mucosal adjuvants. Finally, we discuss future approaches for the development of iNKT cell agonists used as vaccine adjuvants against infectious diseases.


Subject(s)
Adjuvants, Vaccine/pharmacology , Communicable Diseases/immunology , Communicable Diseases/therapy , Natural Killer T-Cells/immunology , Animals , Humans
20.
Oxid Med Cell Longev ; 2022: 2501279, 2022.
Article in English | MEDLINE | ID: mdl-35132346

ABSTRACT

Eukaryotic cells can initiate several distinct self-destruction mechanisms to display essential roles for the homeostasis maintenance, development, and survival of an organism. Pyroptosis, a key response mode in innate immunity, also referred to as caspase-1-dependent proinflammatory programmed necrotic cell death activated by human caspase-1/4/5, or mouse caspase-1/11, plays indispensable roles in response to cytoplasmic insults and immune defense against infectious diseases. These inflammatory caspases are employed by the host to eliminate pathogen infections such as bacteria, viruses, protozoans, and fungi. Gasdermin D requires to be cleaved and activated by these inflammatory caspases to trigger the pyroptosis process. Physiological rupture of cells results in the release of proinflammatory cytokines, the alarmins IL-1ß and IL-18, symbolizing the inflammatory potential of pyroptosis. Moreover, long noncoding RNAs play direct or indirect roles in the upstream of the pyroptosis trigger pathway. Here, we review in detail recently acquired insights into the central roles of inflammatory caspases, inflammasomes, and pyroptosis, as well as the crosstalk between pyroptosis and long noncoding RNAs in mediating infection immunity and pathogen clearance.


Subject(s)
Caspases/metabolism , Communicable Diseases/immunology , Immunity, Innate , Inflammasomes/metabolism , Pyroptosis/immunology , Signal Transduction/immunology , Alarmins/metabolism , Animals , Communicable Diseases/parasitology , Communicable Diseases/virology , Cytokines/metabolism , Host-Pathogen Interactions/immunology , Humans , Mice , Phosphate-Binding Proteins/metabolism , Pore Forming Cytotoxic Proteins/metabolism , RNA, Long Noncoding/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...