Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Prog Mol Biol Transl Sci ; 190(1): 219-276, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36008000

RESUMO

Medical abzymology has made a great contribution to the development of general autoimmunity theory: it has put the autoantibodies (Ab) as the key brick of the theory to the level of physiological functionality by providing such Ab with the ability to catalyze and mediate direct and independent cytotoxic effect on cellular and molecular targets. Natural catalytic autoantibodies (abzymes) while being a pool of canonical Abs and possessing catalytic activity belong to the new group of physiologically active substances whose features and properties are evolutionary consolidated in one functionally active biomolecule. Therefore, further studies on Ab-mediated autoAg degradation and other targeted Ab-mediated proteolysis may provide biomarkers of newer generations and thus a supplementary tool for assessing the disease progression and predicting disability of the patients and persons at risks. This chapter is a summary of current knowledge and prognostic perspectives toward catalytic Abs in autoimmunity and thus some autoimmune clinical cases, their role in pathogenesis, and the exploitation of both whole molecules and their constituent parts in developing highly effective targeted drugs of the future to come, and thus the therapeutic protocols being individualized.


Assuntos
Anticorpos Catalíticos , Autoimunidade , Anticorpos Catalíticos/metabolismo , Autoanticorpos/metabolismo , Biomarcadores , Progressão da Doença , Humanos
2.
N Engl J Med ; 385(14): 1292-1301, 2021 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-34587386

RESUMO

BACKGROUND: Structural birth defects occur in approximately 3% of live births; most such defects lack defined genetic or environmental causes. Despite advances in surgical approaches, pharmacologic prevention remains largely out of reach. METHODS: We queried worldwide databases of 20,248 families that included children with neurodevelopmental disorders and that were enriched for parental consanguinity. Approximately one third of affected children in these families presented with structural birth defects or microcephaly. We performed exome or genome sequencing of samples obtained from the children, their parents, or both to identify genes with biallelic pathogenic or likely pathogenic mutations present in more than one family. After identifying disease-causing variants, we generated two mouse models, each with a pathogenic variant "knocked in," to study mechanisms and test candidate treatments. We administered a small-molecule Wnt agonist to pregnant animals and assessed their offspring. RESULTS: We identified homozygous mutations in WLS, which encodes the Wnt ligand secretion mediator (also known as Wntless or WLS) in 10 affected persons from 5 unrelated families. (The Wnt ligand secretion mediator is essential for the secretion of all Wnt proteins.) Patients had multiorgan defects, including microcephaly and facial dysmorphism as well as foot syndactyly, renal agenesis, alopecia, iris coloboma, and heart defects. The mutations affected WLS protein stability and Wnt signaling. Knock-in mice showed tissue and cell vulnerability consistent with Wnt-signaling intensity and individual and collective functions of Wnts in embryogenesis. Administration of a pharmacologic Wnt agonist partially restored embryonic development. CONCLUSIONS: Genetic variations affecting a central Wnt regulator caused syndromic structural birth defects. Results from mouse models suggest that what we have named Zaki syndrome is a potentially preventable disorder. (Funded by the National Institutes of Health and others.).


Assuntos
Anormalidades Múltiplas/genética , Anormalidades Congênitas/genética , Pleiotropia Genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Mutação , Receptores Acoplados a Proteínas G/genética , Proteínas Wnt/metabolismo , Animais , Modelos Animais de Doenças , Fibroblastos/metabolismo , Técnicas de Introdução de Genes , Genes Recessivos , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Camundongos Transgênicos , Linhagem , Fenótipo , Receptores Acoplados a Proteínas G/metabolismo , Síndrome , Via de Sinalização Wnt
3.
Neuron ; 109(2): 241-256.e9, 2021 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-33220177

RESUMO

Autosomal-recessive cerebellar hypoplasia and ataxia constitute a group of heterogeneous brain disorders caused by disruption of several fundamental cellular processes. Here, we identified 10 families showing a neurodegenerative condition involving pontocerebellar hypoplasia with microcephaly (PCHM). Patients harbored biallelic mutations in genes encoding the spliceosome components Peptidyl-Prolyl Isomerase Like-1 (PPIL1) or Pre-RNA Processing-17 (PRP17). Mouse knockouts of either gene were lethal in early embryogenesis, whereas PPIL1 patient mutation knockin mice showed neuron-specific apoptosis. Loss of either protein affected splicing integrity, predominantly affecting short and high GC-content introns and genes involved in brain disorders. PPIL1 and PRP17 form an active isomerase-substrate interaction, but we found that isomerase activity is not critical for function. Thus, we establish disrupted splicing integrity and "major spliceosome-opathies" as a new mechanism underlying PCHM and neurodegeneration and uncover a non-enzymatic function of a spliceosomal proline isomerase.


Assuntos
Proteínas de Ciclo Celular/genética , Doenças Cerebelares/genética , Microcefalia/genética , Mutação/genética , Peptidilprolil Isomerase/genética , Fatores de Processamento de RNA/genética , Spliceossomos/genética , Sequência de Aminoácidos , Animais , Proteínas de Ciclo Celular/química , Doenças Cerebelares/complicações , Doenças Cerebelares/diagnóstico por imagem , Estudos de Coortes , Feminino , Técnicas de Inativação de Genes/métodos , Células HEK293 , Transtornos Heredodegenerativos do Sistema Nervoso/complicações , Transtornos Heredodegenerativos do Sistema Nervoso/diagnóstico por imagem , Transtornos Heredodegenerativos do Sistema Nervoso/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microcefalia/complicações , Microcefalia/diagnóstico por imagem , Linhagem , Peptidilprolil Isomerase/química , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Fatores de Processamento de RNA/química
4.
Front Pediatr ; 6: 373, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30564562

RESUMO

The illness ME/CFS has been repeatedly tied to infectious agents such as Epstein Barr Virus. Expanding research on the human microbiome now allows ME/CFS-associated pathogens to be studied as interacting members of human microbiome communities. Humans harbor these vast ecosystems of bacteria, viruses and fungi in nearly all tissue and blood. Most well-studied inflammatory conditions are tied to dysbiosis or imbalance of the human microbiome. While gut microbiome dysbiosis has been identified in ME/CFS, microbes and viruses outside the gut can also contribute to the illness. Pathobionts, and their associated proteins/metabolites, often control human metabolism and gene expression in a manner that pushes the body toward a state of illness. Intracellular pathogens, including many associated with ME/CFS, drive microbiome dysbiosis by directly interfering with human transcription, translation, and DNA repair processes. Molecular mimicry between host and pathogen proteins/metabolites further complicates this interference. Other human pathogens disable mitochondria or dysregulate host nervous system signaling. Antibodies and/or clonal T cells identified in patients with ME/CFS are likely activated in response to these persistent microbiome pathogens. Different human pathogens have evolved similar survival mechanisms to disable the host immune response and host metabolic pathways. The metabolic dysfunction driven by these organisms can result in similar clusters of inflammatory symptoms. ME/CFS may be driven by this pathogen-induced dysfunction, with the nature of dysbiosis and symptom presentation varying based on a patient's unique infectious and environmental history. Under such conditions, patients would benefit from treatments that support the human immune system in an effort to reverse the infectious disease process.

5.
Discov Med ; 25(140): 299-308, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30021103

RESUMO

The theory of autoimmunity was developed at a time when the human body was regarded as largely sterile. Antibodies in patients with chronic inflammatory disease could consequently not be tied to persistent human pathogens. The concept of the "autoantibody" was created to reconcile this phenomenon. Today, however, the discovery of the human microbiome has revolutionized our understanding of human biology. Humans are superorganisms that harbor trillions of persistent microbial cells. Indeed, vast human microbiomes have been detected in human tissue and blood. These microbial ecosystems harbor thousands of newly identified bacteria, viruses, and other microorganisms -- most of which can act as pathogens under conditions of immunosuppression. The theory of autoimmunity must be revised to account for the human microbiome. Here, we propose a model in which "autoantibodies" are created in response to chronic, persistent microbiome pathogens. The structural homology (molecular mimicry) between pathogen and host proteins can result in "collateral damage" to surrounding human tissue. This calls for a paradigm shift in autoimmune disease treatment. Immunosuppressive medications palliate inflammatory symptoms at the expense of microbiome health and balance. In contrast, treatments that support the immune system in autoimmune disease could allow patients to target pathogens at the root of the disease process.


Assuntos
Autoanticorpos/imunologia , Autoimunidade , Microbiota , Modelos Imunológicos , Mimetismo Molecular/imunologia , Disbiose/imunologia , Disbiose/microbiologia , Humanos
6.
Discov Med ; 23(124): 51-60, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28245427

RESUMO

An extensive microbiome comprised of bacteria, viruses, bacteriophages, and fungi is now understood to persist in nearly every human body site, including tissue and blood. The genomes of these microbes continually interact with the human genome in order to regulate host metabolism. Many components of this microbiome are capable of both commensal and pathogenic activity. They are additionally able to persist in both 'acute' and chronic forms. Inflammatory conditions historically studied separately (autoimmune, neurological and malignant) are now repeatedly tied to a common trend: imbalance or dysbiosis of these microbial ecosystems. Population-based studies of the microbiome can shed light on this dysbiosis. However, it is the collective activity of the microbiome that drives inflammatory processes via complex microbe-microbe and host-microbe interactions. Many microbes survive as polymicrobial entities in order to evade the immune response. Pathogens in these communities alter their gene expression in ways that promote community-wide virulence. Other microbes persist inside the cells of the immune system, where they directly interfere with host transcription, translation, and DNA repair mechanisms. The numerous proteins and metabolites expressed by these pathogens further dysregulate human gene expression in a manner that promotes imbalance and immunosuppression. Molecular mimicry, or homology between host and microbial proteins, complicates the nature of this interference. When taken together, these microbe-microbe and host-microbe interactions are capable of driving the large-scale failure of human metabolism characteristic of many different inflammatory conditions.


Assuntos
Disbiose/microbiologia , Microbiota/fisiologia , Animais , Disbiose/imunologia , Disbiose/virologia , Microbioma Gastrointestinal/imunologia , Interações Hospedeiro-Patógeno , Humanos , Inflamação/metabolismo
7.
Immunol Res ; 65(1): 129-135, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27412293

RESUMO

Studies in mice have shown that environmental electromagnetic waves tend to suppress the murine immune system with a potency similar to NSAIDs, yet the nature of any Electrosmog effects upon humans remains controversial. Previously, we reported how the human Vitamin-D receptor (VDR) and its ligand, 1,25-dihydroxyvitamin-D (1,25-D), are associated with many chronic inflammatory and autoimmune diseases. We have shown how olmesartan, a drug marketed for mild hypertension, acts as a high-affinity partial agonist for the VDR, and that it seems to reverse disease activity resulting from VDR dysfunction. We here report that structural instability of the activated VDR becomes apparent when observing hydrogen bond behavior with molecular dynamics, revealing that the VDR pathway exhibits a susceptibility to Electrosmog. Further, we note that characteristic modes of instability lie in the microwave frequency range, which is currently populated by cellphone and WiFi communication signals, and that the susceptibility is ligand dependent. A case series of 64 patient-reported outcomes subsequent to use of a silver-threaded cap designed to protect the brain and brain stem from microwave Electrosmog resulted in 90 % reporting "definite" or "strong" changes in their disease symptoms. This is much higher than the 3-5 % rate reported for electromagnetic hypersensitivity in a healthy population and suggests that effective control of environmental Electrosmog immunomodulation may soon become necessary for successful therapy of autoimmune disease.


Assuntos
Doenças Autoimunes , Micro-Ondas , Exposição Ambiental/prevenção & controle , Humanos , Imidazóis/uso terapêutico , Imunomodulação , Receptores de Calcitriol , Tetrazóis/uso terapêutico
8.
Discov Med ; 17(95): 257-65, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24882717

RESUMO

The human body is a superorganism in which thousands of microbial genomes continually interact with the human genome. A range of physical and neurological inflammatory diseases are now associated with shifts in microbiome composition. Seemingly disparate inflammatory conditions may arise from similar disruption of microbiome homeostasis. Intracellular pathogens long associated with inflammatory disease are able to slow the innate immune response by dysregulating activity of the VDR nuclear receptor. This facilitates the ability of other species to gradually accumulate in tissue and blood, where they generate proteins and metabolites that significantly interfere with the body's metabolic processes. The microbes that contribute to this dysfunction are often inherited from family members. Immunosuppressive therapies for inflammatory disease allow pathogens driving these processes to spread with greater ease. In contrast to immunosuppression, treatments that stimulate the immune system seem to allow for reversal of this pathogen-induced genomic dysregulation.


Assuntos
Inflamação/microbiologia , Inflamação/fisiopatologia , Microbiota , Regulação Bacteriana da Expressão Gênica , Genoma Humano , Genômica , Humanos , Imunidade Inata , Metagenoma
9.
Immunol Res ; 56(2-3): 398-412, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23576059

RESUMO

Chronic fatigue syndrome (CFS)/myalgic encephalomyelitis (ME) has long been associated with the presence of infectious agents, but no single pathogen has been reliably identified in all patients with the disease. Recent studies using metagenomic techniques have demonstrated the presence of thousands of microbes in the human body that were previously undetected and unknown to science. More importantly, such species interact together by sharing genes and genetic function within communities. It follows that searching for a singular pathogen may greatly underestimate the microbial complexity potentially driving a complex disease like CFS/ME. Intracellular microbes alter the expression of human genes in order to facilitate their survival. We have put forth a model describing how multiple species-bacterial, viral, and fungal-can cumulatively dysregulate expression by the VDR nuclear receptor in order to survive and thus drive a disease process. Based on this model, we have developed an immunostimulatory therapy that is showing promise inducing both subjective and objective improvement in patients suffering from CFS/ME.


Assuntos
Coinfecção/imunologia , Síndrome de Fadiga Crônica/terapia , Infecções/imunologia , Receptores de Calcitriol/metabolismo , Coinfecção/microbiologia , Coinfecção/terapia , Disbiose , Síndrome de Fadiga Crônica/imunologia , Síndrome de Fadiga Crônica/microbiologia , Regulação da Expressão Gênica/imunologia , Humanos , Imunidade Inata , Imunização , Terapia de Imunossupressão , Infecções/microbiologia , Infecções/terapia , Metagenoma/imunologia , Microbiota/imunologia , Modelos Biológicos , Receptores de Calcitriol/genética
10.
Curr Opin Rheumatol ; 25(2): 234-40, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23370376

RESUMO

PURPOSE OF REVIEW: To demonstrate how dysbiosis of the human microbiome can drive autoimmune disease. RECENT FINDINGS: Humans are superorganisms. The human body harbors an extensive microbiome, which has been shown to differ in patients with autoimmune diagnoses. Intracellular microbes slow innate immune defenses by dysregulating the vitamin D nuclear receptor, allowing pathogens to accumulate in tissue and blood. Molecular mimicry between pathogen and host causes further dysfunction by interfering with human protein interactions. Autoantibodies may well be created in response to pathogens. SUMMARY: The catastrophic failure of human metabolism observed in autoimmune disease results from a common underlying pathogenesis - the successive accumulation of pathogens into the microbiome over time, and the ability of such pathogens to dysregulate gene transcription, translation, and human metabolic processes. Autoimmune diseases are more likely passed in families because of the inheritance of a familial microbiome, rather than Mendelian inheritance of genetic abnormalities. We can stimulate innate immune defenses and allow patients to target pathogens, but cell death results in immunopathology.


Assuntos
Autoimunidade , Metagenoma/imunologia , Adjuvantes Imunológicos/uso terapêutico , Autoanticorpos/biossíntese , Doenças Autoimunes/etiologia , Doenças Autoimunes/microbiologia , Doenças Autoimunes/terapia , Família , Feminino , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Inata , Infecções/imunologia , Infecções/microbiologia , Masculino , Modelos Imunológicos , Mimetismo Molecular/imunologia , Polimorfismo de Nucleotídeo Único , Receptores de Calcitriol/imunologia
11.
Cell Mol Immunol ; 8(3): 213-25, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21278764

RESUMO

Microbes are increasingly being implicated in autoimmune disease. This calls for a re-evaluation of how these chronic inflammatory illnesses are routinely treated. The standard of care for autoimmune disease remains the use of medications that slow the immune response, while treatments aimed at eradicating microbes seek the exact opposite-stimulation of the innate immune response. Immunostimulation is complicated by a cascade of sequelae, including exacerbated inflammation, which occurs in response to microbial death. Over the past 8 years, we have collaborated with American and international clinical professionals to research a model-based treatment for inflammatory disease. This intervention, designed to stimulate the innate immune response, has required a reevaluation of disease progression and amelioration. Paramount is the inherent conflict between palliation and microbicidal efficacy. Increased microbicidal activity was experienced as immunopathology-a temporary worsening of symptoms. Further studies are needed, but they will require careful planning to manage this immunopathology.


Assuntos
Doenças Autoimunes/imunologia , Bactérias/imunologia , Infecções/imunologia , Metagenoma/imunologia , Vírus/imunologia , Doenças Autoimunes/etiologia , Doenças Autoimunes/fisiopatologia , Bactérias/patogenicidade , Progressão da Doença , Humanos , Imunidade Inata , Imunização , Infecções/complicações , Infecções/fisiopatologia , Vírus/patogenicidade
12.
Ann N Y Acad Sci ; 1173: 252-9, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19758159

RESUMO

Researchers have noted that the incidence of autoimmune diseases, such as Hashimoto's thyroiditis, is markedly higher in women than in men, but to date the reason for this disparity has been unclear. The vitamin D nuclear receptor (VDR) is expressed in the human cycling endometrium. Because the VDR controls expression of the cathelicidin and beta-defensin antimicrobial peptides (AmPs), dysregulation of the receptor greatly compromises the innate immune response. Increasing evidence indicates the presence of a chronic, intraphagocytic, metagenomic microbiota in patients with autoimmune disease that may survive by dysregulating the VDR. VDR dysregulation, in turn, prevents the breakdown of the active vitamin D metabolite 1,25-hydroxyvitamin D (1,25-D) by CYP24. In silico data suggest that when 1,25-D rises above its normal range, it binds the alpha/beta thyroid receptors, the glucocorticoid receptor (GCR), and the androgen receptor (AR), displacing their native ligands and causing an array of hormonal imbalances. If T3 is displaced from alpha-thyroid, thyroiditis may result. Because the VDR, GCR, and AR also express multiple families of AmPs, expression of these natural antibiotics further wanes in response to dysregulation by 1,25-D. The end result is a system-wide drop in AmP expression that may allow pathogens to spread with greater ease. Because women have an extra site of VDR expression in the endometrium, the drop in AmP expression associated with nuclear receptor dysregulation may disproportionately affect them. This would cause women to accumulate higher bacterial loads than their male counterparts, particularly during early pregnancy when 1,25-D levels rise by 40%.


Assuntos
Doenças Autoimunes/fisiopatologia , Núcleo Celular/metabolismo , Receptores de Calcitriol/fisiologia , Peptídeos Catiônicos Antimicrobianos/metabolismo , Doenças Autoimunes/metabolismo , Sítios de Ligação , Calcitriol/metabolismo , Endométrio/metabolismo , Feminino , Humanos , Imunidade Inata/fisiologia , Ligantes , Masculino , Gravidez , Receptores de Calcitriol/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores Citoplasmáticos e Nucleares/fisiologia , Fatores Sexuais , Esteroide Hidroxilases/metabolismo , Vitamina D3 24-Hidroxilase , beta-Defensinas/metabolismo , Catelicidinas
13.
Autoimmun Rev ; 8(8): 677-81, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19393196

RESUMO

Studies of autoimmune disease have focused on the characteristics of the identifiable antibodies. But as our knowledge of the genes associated with the disease states expands, we understand that humans must be viewed as superorganisms in which a plethora of bacterial genomes - a metagenome - work in tandem with our own. The NIH has estimated that 90% of the cells in Homo sapiens are microbial and not human in origin. Some of these microbes create metabolites that interfere with the expression of genes associated with autoimmune disease. Thus, we must re-examine how human gene transcription is affected by the plethora of microbial metabolites. We can no longer assume that antibodies generated in autoimmune disease are created solely as autoantibodies to human DNA. Evidence is now emerging that the human microbiota accumulates during a lifetime, and a variety of persistence mechanisms are coming to light. In one model, obstruction of VDR nuclear-receptor-transcription prevents the innate immune system from making key antimicrobials, allowing the microbes to persist. Genes from these microbes must necessarily impact disease progression. Recent efforts to decrease this VDR-perverting microbiota in patients with autoimmune disease have resulted in reversal of autoimmune processes. As the NIH Human Microbiome Project continues to better characterize the human metagenome, new insights into autoimmune pathogenesis are beginning to emerge.


Assuntos
Doenças Autoimunes/microbiologia , Bactérias/metabolismo , Genômica , Metaboloma/genética , Metabolômica , Metagenoma/genética , Ácidos Alcanossulfônicos/metabolismo , Autoanticorpos/imunologia , Doenças Autoimunes/genética , Doenças Autoimunes/metabolismo , Bactérias/genética , DNA Bacteriano/metabolismo , Humanos , Imunidade Inata
14.
Autoimmun Rev ; 8(8): 639-44, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19393200

RESUMO

Early studies on vitamin D showed promise that various forms of the "vitamin" may be protective against chronic disease, yet systematic reviews and longer-term studies have failed to confirm these findings. A number of studies have suggested that patients with autoimmune diagnoses are deficient in 25-hydroxyvitamin D (25-D) and that consuming greater quantities of vitamin D, which further elevates 25 D levels, alleviates autoimmune disease symptoms. Some years ago, molecular biology identified 25 D as a secosteroid. Secosteroids would typically be expected to depress inflammation, which is in line with the reports of symptomatic improvement. The simplistic first-order mass-action model used to guide the early vitamin studies is now giving way to a more complex description of action. When active, the Vitamin D nuclear receptor (VDR) affects transcription of at least 913 genes and impacts processes ranging from calcium metabolism to expression of key antimicrobial peptides. Additionally, recent research on the Human Microbiome shows that bacteria are far more pervasive than previously thought, increasing the possibility that autoimmune disease is bacterial in origin. Emerging molecular evidence suggests that symptomatic improvements among those administered vitamin D is the result of 25-D's ability to temper bacterial-induced inflammation by slowing VDR activity. While this results in short-term palliation, persistent pathogens that may influence disease progression, proliferate over the long-term.


Assuntos
Doenças Autoimunes/imunologia , Neoplasias/imunologia , Receptores de Calcitriol/imunologia , Deficiência de Vitamina D/imunologia , Vitamina D/análogos & derivados , Doenças Autoimunes/metabolismo , Bactérias/imunologia , Bactérias/metabolismo , Humanos , Neoplasias/metabolismo , Receptores de Calcitriol/metabolismo , Vitamina D/imunologia , Vitamina D/metabolismo , Deficiência de Vitamina D/metabolismo
15.
Bioessays ; 30(2): 173-82, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18200565

RESUMO

The US FDA currently encourages the addition of vitamin D to milk and cereals, with the aim of reducing rickets in children and osteoporosis in adults. However, vitamin D not only regulates the expression of genes associated with calcium homeostasis, but also genes associated with cancers, autoimmune disease, and infection. It does this by controlling the activation of the vitamin D receptor (VDR), a type 1 nuclear receptor and DNA transcription factor. Molecular biology is rapidly coming to an understanding of the multiplicity of roles played by the VDR, but clinical medicine is having difficulty keeping up with the pace of change. For example, the FDA recently proposed a rule change that will encourage high levels of vitamin D to be added to even more foods, so that the manufacturers can claim those foods "reduce the risk of osteoporosis". The FDA docket does not review one single paper detailing the transcriptional activity of vitamin D, even though, on average, one new paper a day is being published on that topic. Nor do they review whether widespread supplementation with vitamin D, an immunomodulatory secosteroid, might predispose the population to immune dysfunction. This BioEssay explores how lifelong supplementation of the food chain with vitamin D might well be contributing to the current epidemics of obesity and chronic disease.


Assuntos
Tomada de Decisões , United States Food and Drug Administration , Vitamina D/efeitos adversos , Cálcio/metabolismo , Humanos , Obesidade/induzido quimicamente , Fatores de Risco , Estados Unidos , Vitamina D/metabolismo
17.
Theor Biol Med Model ; 3: 1, 2006 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-16403216

RESUMO

BACKGROUND: There have been indications that common Angiotensin Receptor Blockers (ARBs) may be exerting anti-inflammatory actions by directly modulating the immune system. We decided to use molecular modelling to rapidly assess which of the potential targets might justify the expense of detailed laboratory validation. We first studied the VDR nuclear receptor, which is activated by the secosteroid hormone 1,25-dihydroxyvitamin-D. This receptor mediates the expression of regulators as ubiquitous as GnRH (Gonadatrophin hormone releasing hormone) and the Parathyroid Hormone (PTH). Additionally we examined Peroxisome Proliferator-Activated Receptor Gamma (PPARgamma), which affects the function of phagocytic cells, and the C-CChemokine Receptor, type 2b, (CCR2b), which recruits monocytes to the site of inflammatory immune challenge. RESULTS: Telmisartan was predicted to strongly antagonize (Ki asymptotically equal to 0.04 nmol) the VDR. The ARBs Olmesartan, Irbesartan and Valsartan (Ki asymptotically equal to10 nmol) are likely to be useful VDR antagonists at typical in-vivo concentrations. Candesartan (Ki asymptotically equal to 30 nmol) and Losartan (Ki asymptotically equal to 70 nmol) may also usefully inhibit the VDR. Telmisartan is a strong modulator of PPARgamma (Ki asymptotically equal to 0.3 nmol), while Losartan (Ki asymptotically equal to 3 nmol), Irbesartan (Ki asymptotically equal to 6 nmol), Olmesartan and Valsartan (Ki asymptotically equal to 12 nmol) also seem likely to have significant PPAR modulatory activity. Olmesartan and Irbesartan (Ki asymptotically equal to 9 nmol) additionally act as antagonists of a theoretical model of CCR2b. Initial validation of this CCR2b model was performed, and a proposed model for the Angiotensin II Type1 receptor (AT2R1) has been presented. CONCLUSION: Molecular modeling has proven valuable to generate testable hypotheses concerning receptor/ligand binding and is an important tool in drug design. ARBs were designed to act as antagonists for AT2R1, and it was not surprising to discover their affinity for the structurally similar CCR2b. However, this study also found evidence that ARBs modulate the activation of two key nuclear receptors-VDR and PPARgamma. If our simulations are confirmed by experiment, it is possible that ARBs may become useful as potent anti-inflammatory agents, in addition to their current indication as cardiovascular drugs.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Antagonistas de Receptores de Angiotensina , PPAR gama/metabolismo , Receptores de Calcitriol/metabolismo , Receptores de Quimiocinas/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Simulação por Computador , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Receptores de Angiotensina/metabolismo , Receptores CCR2
18.
Autoimmun Rev ; 3(4): 295-300, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15246025

RESUMO

From time to time there have been reports of autoimmune disease succumbing to tetracycline antibiotics, but many have assumed this was due to coincidence, or to some ill-defined 'anti-inflammatory property' of the tetracyclines. But now the inflammation of sarcoidosis has succumbed to antibiotics in two independent studies. This review examines the cell wall deficient (antibiotic resistant) bacteria which have been found in tissue from patients with sarcoidosis. It examines how such bacteria can infect the phagocytes of the immune system, and how they may therefore be responsible for not only sarcoid inflammation, but also for other autoimmune disease. Proof positive of a bacterial pathogenesis for Sarcoidosis includes not only the demonstrated ability of these studies to put the disease into remission, but also the severity of Jarisch-Herxheimer shock resulting from endotoxin release as the microbes are killed. Studies delineating the hormone responsible for phagocyte differentiation in the Th1 immune response, 1,25-dihydroxyvitamin D, are discussed, and its utility as a marker of Th1 immune inflammation is reviewed. Finally, data showing that the behavior of this hormone is also aberrant in rheumatoid arthritis, systemic lupus erythematosus, and Parkinson's, raise the possibility that these diseases may also have a CWD bacterial pathogenesis.


Assuntos
Antibacterianos/uso terapêutico , Sarcoidose/tratamento farmacológico , Tetraciclinas/uso terapêutico , Vitamina D/análogos & derivados , Animais , Artrite Reumatoide/tratamento farmacológico , Doenças Autoimunes/tratamento farmacológico , Doenças Autoimunes/etiologia , Bactérias , Doxiciclina/uso terapêutico , Previsões , Humanos , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Minociclina/uso terapêutico , Doença de Parkinson/tratamento farmacológico , Fagócitos/patologia , Sarcoidose/etiologia , Sarcoidose/metabolismo , Sarcoidose/patologia , Prevenção Secundária , Vitamina D/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA