Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Z Rheumatol ; 83(2): 160-168, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38240817

ABSTRACT

OBJECTIVE: Rheumatoid arthritis-associated interstitial lung disease (RA-ILD) is a major driver of premature mortality in patients with rheumatoid arthritis (RA). Detection of RA-ILD is crucial but requires awareness among the treating physicians. To date, however, there is no international recommendation concerning screening for ILD in RA patients. METHODS: After a systematic literature review, the modified Delphi technique in combination with the nominal group technique was used to provide a Delphi consensus statement elaborated by an expert panel of pneumonologists, rheumatologists, and a radiologist. Based on the available evidence, several clusters of questions were defined and discussed until consent was reached. RESULTS: A screening algorithm for ILD in patients with RA based on clinical signs, respiratory symptoms, and risk factors has been developed. Further, the recommendations address diagnostic tools for RA-ILD and the follow-up of RA patients qualifying for ILD screening.


Subject(s)
Arthritis, Rheumatoid , Lung Diseases, Interstitial , Humans , Arthritis, Rheumatoid/complications , Arthritis, Rheumatoid/diagnosis , Arthritis, Rheumatoid/drug therapy , Lung Diseases, Interstitial/diagnosis , Lung Diseases, Interstitial/etiology , Risk Factors
3.
Z Rheumatol ; 82(2): 163-174, 2023 Mar.
Article in German | MEDLINE | ID: mdl-36342525

ABSTRACT

This nationwide Austrian consensus statement summarizes the recommendations on the management of latent tuberculosis by treatment with biologic and targeted synthetic DMARDs. The essential questions with respect to screening and preventive treatment were discussed by experts from the disciplines of rheumatology, pneumology, infectious diseases, dermatology and gastroenterology, based on the available data, and then a joint consensus was formed by agreement. This involved a differentiated discussion on the various forms of treatment, and clear recommendations were formulated.


Subject(s)
Antirheumatic Agents , Arthritis, Rheumatoid , Biological Products , Dermatology , Gastroenterology , Latent Tuberculosis , Pulmonary Medicine , Rheumatology , Humans , Antirheumatic Agents/therapeutic use , Austria , Arthritis, Rheumatoid/diagnosis , Arthritis, Rheumatoid/drug therapy , Latent Tuberculosis/drug therapy , Biological Products/adverse effects
4.
Wien Klin Wochenschr ; 134(21-22): 751-765, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36036323

ABSTRACT

This publication provides a thorough analysis of the most relevant topics concerning the management of latent tuberculosis when using biologic and targeted synthetic Disease Modifying Antirheumatic Drugs (DMARDs) by a multidisciplinary, select committee of Austrian physicians. The committee includes members of the Austrian Societies for Rheumatology and Rehabilitation, Pulmonology, Infectiology, Dermatology and Gastroenterology. Consensus was reached on issues regarding screening and treatment of latent tuberculosis and includes separate recommendations for each biologic and targeted synthetic DMARD.


Subject(s)
Antirheumatic Agents , Arthritis, Rheumatoid , Biological Products , Dermatology , Gastroenterology , Latent Tuberculosis , Pulmonary Medicine , Rheumatology , Humans , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/diagnosis , Arthritis, Rheumatoid/drug therapy , Latent Tuberculosis/diagnosis , Latent Tuberculosis/drug therapy , Austria , Biological Products/therapeutic use
5.
J Clin Invest ; 132(13)2022 07 01.
Article in English | MEDLINE | ID: mdl-35653190

ABSTRACT

Mitochondrial proteostasis, regulated by the mitochondrial unfolded protein response (UPRmt), is crucial for maintenance of cellular functions and survival. Elevated oxidative and proteotoxic stress in mitochondria must be attenuated by the activation of a ubiquitous UPRmt to promote prostate cancer (PCa) growth. Here we show that the 2 key components of the UPRmt, heat shock protein 60 (HSP60, a mitochondrial chaperonin) and caseinolytic protease P (ClpP, a mitochondrial protease), were required for the development of advanced PCa. HSP60 regulated ClpP expression via c-Myc and physically interacted with ClpP to restore mitochondrial functions that promote cancer cell survival. HSP60 maintained the ATP-producing functions of mitochondria, which activated the ß-catenin pathway and led to the upregulation of c-Myc. We identified a UPRmt inhibitor that blocked HSP60's interaction with ClpP and abrogated survival signaling without altering HSP60's chaperonin function. Disruption of HSP60-ClpP interaction with the UPRmt inhibitor triggered metabolic stress and impeded PCa-promoting signaling. Treatment with the UPRmt inhibitor or genetic ablation of Hsp60 inhibited PCa growth and progression. Together, our findings demonstrate that the HSP60-ClpP-mediated UPRmt is essential for prostate tumorigenesis and the HSP60-ClpP interaction represents a therapeutic vulnerability in PCa.


Subject(s)
Chaperonin 60 , Prostatic Neoplasms , Animals , Chaperonin 60/genetics , Chaperonin 60/metabolism , Humans , Male , Mice , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Peptide Hydrolases/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Unfolded Protein Response
6.
Mucosal Immunol ; 15(4): 595-604, 2022 04.
Article in English | MEDLINE | ID: mdl-35534699

ABSTRACT

The intestinal epithelium represents the most regenerative tissue in the human body, located in proximity to the dense and functionally diverse microbial milieu of the microbiome. Episodes of tissue injury and incomplete healing of the intestinal epithelium are a prerequisite for immune reactivation and account for recurrent, chronically progressing phenotypes of inflammatory bowel diseases (IBD). Mitochondrial dysfunction and associated changes in intestinal epithelial functions are emerging concepts in the pathogenesis of IBD, suggesting impaired metabolic flexibility of epithelial cells affects the regenerative capacity of the intestinal tissue. Next to rendering the intestinal mucosa susceptible to inflammatory triggers, metabolic reprogramming of the epithelium is implicated in shaping adverse microbial environments. In this review, we introduce the concept of "metabolic injury" as a cell autonomous mechanism of tissue wounding in response to mitochondrial perturbation. Furthermore, we highlight epithelial metabolism as intersection of microbiome, immune cells and epithelial regeneration.


Subject(s)
Inflammatory Bowel Diseases , Microbiota , Dysbiosis/complications , Epithelial Cells/metabolism , Humans , Intestinal Mucosa
7.
Pharmaceutics ; 13(8)2021 Aug 17.
Article in English | MEDLINE | ID: mdl-34452240

ABSTRACT

The intestinal epithelium critically contributes to oral bioavailability of drugs by constituting an important site for drug absorption and metabolism. In particular, intestinal epithelial cells (IEC) actively serve as gatekeepers of drug and nutrient availability. IECs' transport processes and metabolism are interrelated to the whole-body metabolic state and represent potential points of origin as well as therapeutic targets for a variety of diseases. Human intestinal organoids represent a superior model of the intestinal epithelium, overcoming limitations of currently used in vitro models. Caco-2 cells or rodent explant models face drawbacks such as their cancer and non-human origin, respectively, but are commonly used to study intestinal nutrient absorption, enterocyte metabolism and oral drug bioavailability, despite poorly correlative data. In contrast, intestinal organoids allow investigating distinct aspects of bioavailability including spatial resolution of transport, inter-individual differences and high-throughput screenings. As several countries have already developed strategic roadmaps to phase out animal experiments for regulatory purposes, intestinal organoid culture and organ-on-a-chip technology in combination with in silico approaches are roads to go in the preclinical and regulatory setup and will aid implementing the 3Rs (reduction, refinement and replacement) principle in basic science.

8.
Life Sci ; 267: 118974, 2021 Feb 15.
Article in English | MEDLINE | ID: mdl-33385407

ABSTRACT

AIM: We aimed to determine whether the sodium/glucose cotransporter family member SGLT3, a proposed glucose sensor, is expressed in the intestine and/or kidney, and if its expression is altered in mouse models of obesity and in humans before and after weight-loss surgery. MAIN METHODS: We used in-situ hybridization and quantitative PCR to determine whether the Sglt3 isoforms 3a and 3b were expressed in the intestine and kidney of C57, leptin-deficient ob/ob, and diabetic BTBR ob/ob mice. Western blotting and immunohistochemistry were also used to assess SGLT3 protein levels in jejunal biopsies from obese patients before and after weight-loss Roux-en-Y gastric bypass surgery (RYGB), and in lean healthy controls. KEY FINDINGS: Sglt3a/3b mRNA was detected in the small intestine (duodenum, jejunum and ileum), but not in the large intestine or kidneys of mice. Both isoforms were detected in epithelial cells (confirmed using intestinal organoids). Expression of Sglt3a/3b mRNA in duodenum and jejunum was significantly lower in ob/ob and BTBR ob/ob mice than in normal-weight littermates. Jejunal SGLT3 protein levels in aged obese patients before RYGB were lower than in lean individuals, but substantially upregulated 6 months post-RYGB. SIGNIFICANCE: Our study shows that Sglt3a/3b is expressed primarily in epithelial cells of the small intestine in mice. Furthermore, we observed an association between intestinal mRNA Sglt3a/3b expression and obesity in mice, and between jejunal SGLT3 protein levels and obesity in humans. Further studies are required to determine the possible role of SGLT3 in obesity.


Subject(s)
Obesity/metabolism , Sodium-Glucose Transport Proteins/genetics , Adult , Animals , Disease Models, Animal , Down-Regulation , Female , Gastric Bypass , Gene Expression , Humans , Insulin/metabolism , Insulin Resistance , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Jejunum/metabolism , Leptin/deficiency , Leptin/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Obesity/genetics , Protein Isoforms , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sodium-Glucose Transport Proteins/biosynthesis , Sodium-Glucose Transport Proteins/metabolism , Transcriptome , Weight Loss
9.
Front Bioeng Biotechnol ; 8: 577656, 2020.
Article in English | MEDLINE | ID: mdl-33015026

ABSTRACT

Intestinal transport and sensing processes and their interconnection to metabolism are relevant to pathologies such as malabsorption syndromes, inflammatory diseases, obesity and type 2 diabetes. Constituting a highly selective barrier, intestinal epithelial cells absorb, metabolize, and release nutrients into the circulation, hence serving as gatekeeper of nutrient availability and metabolic health for the whole organism. Next to nutrient transport and sensing functions, intestinal transporters including peptide transporter 1 (PEPT1) are involved in the absorption of drugs and prodrugs, including certain inhibitors of angiotensin-converting enzyme, protease inhibitors, antivirals, and peptidomimetics like ß-lactam antibiotics. Here, we verify the applicability of 3D organoids for in vitro investigation of intestinal biochemical processes related to transport and metabolism of nutrients and drugs. Establishing a variety of methodologies including illustration of transporter-mediated nutrient and drug uptake and metabolomics approaches, we highlight intestinal organoids as robust and reliable tool in this field of research. Currently used in vitro models to study intestinal nutrient absorption, drug transport and enterocyte metabolism, such as Caco-2 cells or rodent explant models are of limited value due to their cancer and non-human origin, respectively. Particularly species differences result in poorly correlative data and findings obtained in these models cannot be extrapolated reliably to humans, as indicated by high failure rates in drug development pipelines. In contrast, human intestinal organoids represent a superior model of the intestinal epithelium and might help to implement the 3Rs (Reduction, Refinement and Replacement) principle in basic science as well as the preclinical and regulatory setup.

10.
Gut ; 69(11): 1939-1951, 2020 11.
Article in English | MEDLINE | ID: mdl-32111634

ABSTRACT

OBJECTIVE: Reduced Paneth cell (PC) numbers are observed in inflammatory bowel diseases and impaired PC function contributes to the ileal pathogenesis of Crohn's disease (CD). PCs reside in proximity to Lgr5+ intestinal stem cells (ISC) and mitochondria are critical for ISC-renewal and differentiation. Here, we characterise ISC and PC appearance under inflammatory conditions and describe the role of mitochondrial function for ISC niche-maintenance. DESIGN: Ileal tissue samples from patients with CD, mouse models for mitochondrial dysfunction (Hsp60Δ/ΔISC) and CD-like ileitis (TNFΔARE), and intestinal organoids were used to characterise PCs and ISCs in relation to mitochondrial function. RESULTS: In patients with CD and TNFΔARE mice, inflammation correlated with reduced numbers of Lysozyme-positive granules in PCs and decreased Lgr5 expression in crypt regions. Disease-associated changes in PC and ISC appearance persisted in non-inflamed tissue regions of patients with CD and predicted the risk of disease recurrence after surgical resection. ISC-specific deletion of Hsp60 and inhibition of mitochondrial respiration linked mitochondrial function to the aberrant PC phenotype. Consistent with reduced stemness in vivo, crypts from inflamed TNFΔARE mice fail to grow into organoids ex vivo. Dichloroacetate-mediated inhibition of glycolysis, forcing cells to shift to mitochondrial respiration, improved ISC niche function and rescued the ability of TNFΔARE mice-derived crypts to form organoids. CONCLUSION: We provide evidence that inflammation-associated mitochondrial dysfunction in the intestinal epithelium triggers a metabolic imbalance, causing reduced stemness and acquisition of a dysfunctional PC phenotype. Blocking glycolysis might be a novel drug target to antagonise PC dysfunction in the pathogenesis of CD.


Subject(s)
Crohn Disease/etiology , Crohn Disease/pathology , Mitochondria/physiology , Paneth Cells/pathology , Stem Cells/cytology , Animals , Cell Differentiation , Disease Models, Animal , Humans , Mice , Recurrence , Stem Cell Niche
11.
Front Cell Dev Biol ; 8: 602814, 2020.
Article in English | MEDLINE | ID: mdl-33469536

ABSTRACT

Mitochondrial metabolism, dynamics, and stress responses in the intestinal stem cell niche play a pivotal role in regulating intestinal epithelial cell homeostasis, including self-renewal and differentiation. In addition, mitochondria are increasingly recognized for their involvement in sensing the metabolic environment and their capability of integrating host and microbial-derived signals. Gastrointestinal diseases such as inflammatory bowel diseases and colorectal cancer are characterized by alterations of intestinal stemness, the microbial milieu, and mitochondrial metabolism. Thus, mitochondrial function emerges at the interface of determining health and disease, and failure to adapt mitochondrial function to environmental cues potentially results in aberrant tissue responses. A mechanistic understanding of the underlying role of mitochondrial fitness in intestinal pathologies is still in its infancy, and therapies targeting mitochondrial (dys)function are currently lacking. This review discusses mitochondrial signaling and metabolism in intestinal stem cells and Paneth cells as critical junction translating host- and microbe-derived signals into epithelial responses. Consequently, we propose mitochondrial fitness as a hallmark for intestinal epithelial cell plasticity, determining the regenerative capacity of the epithelium.

12.
Microbiome ; 6(1): 134, 2018 08 02.
Article in English | MEDLINE | ID: mdl-30071904

ABSTRACT

BACKGROUND: As the gut microbiota contributes to metabolic health, it is important to determine specific diet-microbiota interactions that influence host metabolism. Bile acids and dietary fat source can alter phenotypes of diet-induced obesity, but the interplay with intestinal microorganisms is unclear. Here, we investigated metabolic consequences of diets enriched in primary bile acids with or without addition of lard or palm oil, and studied gut microbiota structure and functions in mice. RESULTS: In combination with bile acids, dietary lard fed to male C57BL/6N mice for a period of 8 weeks enhanced fat mass accumulation in colonized, but not in germ-free mice when compared to palm oil. This was associated with impaired glucose tolerance, lower fasting insulin levels, lower counts of enteroendocrine cells, fatty liver, and elevated amounts of hepatic triglycerides, cholesteryl esters, and monounsaturated fatty acids. Lard- and bile acid-fed mice were characterized by shifts in dominant gut bacterial communities, including decreased relative abundances of Lachnospiraceae and increased occurrence of Desulfovibrionaceae and the species Clostridium lactatifermentans and Flintibacter butyricus. Metatranscriptomic analysis revealed shifts in microbial functions, including lipid and amino acid metabolism. CONCLUSIONS: Caution is required when interpreting data from diet-induced obesity models due to varying effects of dietary fat source. Detrimental metabolic consequences of a diet enriched with lard and primary bile acids were dependent on microbial colonization of the host and were linked to hepatic lipid rearrangements and to alterations of dominant bacterial communities in the cecum.


Subject(s)
Bacteria/classification , Bile Acids and Salts/analysis , Diet, High-Fat/adverse effects , Gastrointestinal Microbiome/drug effects , Obesity/chemically induced , Amino Acids/metabolism , Animals , Bacteria/drug effects , Bacteria/genetics , DNA, Bacterial/genetics , DNA, Ribosomal/genetics , Dietary Fats/adverse effects , Gene Expression Profiling , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , Palm Oil/adverse effects , Phylogeny , RNA, Ribosomal, 16S/genetics
13.
Gastroenterology ; 155(5): 1539-1552.e12, 2018 11.
Article in English | MEDLINE | ID: mdl-30063920

ABSTRACT

BACKGROUND & AIMS: Activating transcription factor 6 (ATF6) regulates endoplasmic reticulum stress. We studied whether ATF6 contributes to the development of colorectal cancer (CRC) using tissue from patients and transgenic mice. METHODS: We analyzed data from 541 patients with CRC in The Cancer Genome Atlas database for genetic variants and aberrant expression levels of unfolded protein response genes. Findings were validated in a cohort of 83 patients with CRC in Germany. We generated mice with intestinal epithelial cell-specific expression of the active form of Atf6 (nATF6IEC) from 2 alleles (homozygous), mice with expression of nATF6IEC from 1 allele (heterozygous), and nATF6IECfl/fl mice (controls). All nATF6IEC mice were housed under either specific-pathogen-free or germ-free conditions. Cecal microbiota from homozygous nATF6IEC mice or control mice was transferred into homozygous nATF6IEC mice or control mice. nATF6IEC mice were crossed with mice with disruptions in the myeloid differentiation primary response gene 88 and toll-like receptor adaptor molecule 1 gene (Myd88/Trif-knockout mice). Intestinal tissues were collected from mice and analyzed by histology, immunohistochemistry, immunoblots, gene expression profiling of unfolded protein response and inflammatory genes, array-based comparative genome hybridization, and 16S ribosomal RNA gene sequencing. RESULTS: Increased expression of ATF6 was associated with reduced disease-free survival times of patients with CRC. Homozygous nATF6IEC mice developed spontaneous colon adenomas at 12 weeks of age. Compared with controls, homozygous nATF6IEC mice had changes in the profile of their cecal microbiota, increased proliferation of intestinal epithelial cells, and loss of the mucus barrier-all preceding tumor formation. These mice had increased penetration of bacteria into the inner mucus layer and activation of signal transducer and activator of transcription 3, yet inflammation was not observed at the pretumor or tumor stages. Administration of antibiotics to homozygous nATF6IEC mice greatly reduced tumor incidence, and germ-free housing completely prevented tumorigenesis. Analysis of nATF6IEC MyD88/TRIF-knockout mice showed that tumor initiation and growth required MyD88/TRIF-dependent activation of signal transducer and activator of transcription 3. Transplantation of cecal microbiota from nATF6IEC mice and control mice, collected before tumor formation, caused tumor formation in ex-germ-free nATF6IEC mice. CONCLUSIONS: In patients with CRC, ATF6 was associated with reduced time of disease-free survival. In studies of nATF6IEC mice, we found sustained intestinal activation of ATF6 in the colon to promote dysbiosis and microbiota-dependent tumorigenesis.


Subject(s)
Activating Transcription Factor 6/physiology , Colorectal Neoplasms/etiology , Dysbiosis/etiology , Immunity, Innate , Intestines/microbiology , Adaptor Proteins, Vesicular Transport/physiology , Animals , Colorectal Neoplasms/mortality , Disease Progression , Humans , Mice , Myeloid Differentiation Factor 88/physiology , STAT3 Transcription Factor/physiology , Toll-Like Receptors/physiology , Unfolded Protein Response
14.
Nat Rev Gastroenterol Hepatol ; 15(8): 497-516, 2018 08.
Article in English | MEDLINE | ID: mdl-29844587

ABSTRACT

The intestinal epithelium is a multicellular interface in close proximity to a dense microbial milieu that is completely renewed every 3-5 days. Pluripotent stem cells reside at the crypt, giving rise to transient amplifying cells that go through continuous steps of proliferation, differentiation and finally anoikis (a form of programmed cell death) while migrating upwards to the villus tip. During these cellular transitions, intestinal epithelial cells (IECs) possess distinct metabolic identities reflected by changes in mitochondrial activity. Mitochondrial function emerges as a key player in cell fate decisions and in coordinating cellular metabolism, immunity, stress responses and apoptosis. Mediators of mitochondrial signalling include molecules such as ATP and reactive oxygen species and interrelate with pathways such as the mitochondrial unfolded protein response (MT-UPR) and AMP kinase signalling, in turn affecting cell cycle progression and stemness. Alterations in mitochondrial function and MT-UPR activation are integral aspects of pathologies, including IBD and cancer. Mitochondrial signalling and concomitant changes in metabolism contribute to intestinal homeostasis and regulate IEC dedifferentiation-differentiation programmes in the context of diseases, suggesting that mitochondrial function as a cellular checkpoint critically contributes to disease outcome. This Review highlights mitochondrial function and MT-UPR signalling in epithelial cell stemness, differentiation and lineage commitment and illustrates mitochondrial function in intestinal diseases.


Subject(s)
Epithelial Cells/physiology , Homeostasis/physiology , Intestinal Mucosa/physiology , Mitochondria/physiology , Bile Acids and Salts/physiology , Cell Proliferation/physiology , Fatty Acids, Volatile/physiology , Humans , Intestinal Mucosa/cytology , Intestinal Neoplasms/etiology , Intestinal Neoplasms/physiopathology , Irritable Bowel Syndrome/etiology , Irritable Bowel Syndrome/physiopathology , Reactive Oxygen Species/metabolism , Receptors, Pattern Recognition/physiology , Signal Transduction/physiology , Stem Cells/physiology , Unfolded Protein Response/physiology
15.
Endocr Dev ; 32: 49-73, 2017.
Article in English | MEDLINE | ID: mdl-28873384

ABSTRACT

Enteroendocrine cells (EEC) have been studied extensively for their ability to regulate gastrointestinal motility and insulin release by secretion of peptide hormones. In particular, the L cell-derived incretin glucagon-like peptide 1 has gained enormous attention due to its insulinotropic action and relevance in the treatment of type 2 diabetes. Yet, accumulating data indicates a critical role for EEC and incretins in metabolic adaptation and in orchestrating immune responses beyond blood glucose control. EEC actively sense the lamina propria and luminal environment including the microbiota via receptors and transporters, subsequently mediating signals by secreting hormones and cytokines. Data indicate that immune cells and cytokine-mediated signaling impacts EEC numbers and function during infection and chronic inflammation of the gut, suggesting EEC not only to play a role in these pathologies but also being a target of inflammatory processes. This review presents data on the interrelation of incretins and inflammatory signaling. It focuses on the impact of intestinal inflammation, in particular inflammatory bowel disease, on EEC and the potential role of EEC and incretins in these pathologies. Furthermore, it highlights endoplasmic reticulum unfolded protein response, cytokines and the intestinal microbiota as possible targets of inflammatory and EEC signaling.


Subject(s)
Enteroendocrine Cells/immunology , Enteroendocrine Cells/physiology , Gastrointestinal Diseases/physiopathology , Incretins/physiology , Animals , Gastrointestinal Diseases/immunology , Gastrointestinal Hormones/physiology , Gastrointestinal Microbiome , Gastrointestinal Motility/physiology , Glucagon-Like Peptide 1/metabolism , Humans , Inflammatory Bowel Diseases , Insulin/metabolism , Insulin Secretion
16.
Medicine (Baltimore) ; 96(11): e6344, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28296767

ABSTRACT

Rheumatoid arthritis (RA) is a chronic and systemic inflammatory disorder. Conventional radiography, a widely available and cost-effective examination method, remains the standard of reference for the detection and quantification of joint involvement in RA. Fractal dimension (FD) of the trabecular bone structure has been proven to correlate with the bone's physical properties. The present study was designed to use fractal analysis to validate radiograph changes in the hand of RA patients.This study retrospectively evaluated the hand radiographs of 108 subjects. Fifty-four patients were suffering from RA, of which 18 were men and 36 were women. Their ages ranged from 25 to 90 years. The hand radiographs of 54 healthy patients, 18 men and 36 women (age range 23-88 years), were used as the control group. Bone structure value (BSV) is a critical parameter for the assessment and analysis of bone microarchitecture. The BSVs were calculated over the fractal dimension using the Brownian motion.The BSV calculated for ROI showed a significant difference in ROI5 (0.210 ±â€Š0.045), ROI6 (0.186 ±â€Š0.066), and ROI11 (0.201 ±â€Š0.056) in patients with RA, in comparison to the CG (P < 0.05). A significant correlation was observed between anti-CCP and ROI4, ROI5, ROI6, ROI9, and ROI12 in seropositive RA patients (post hoc test (Bonferroni) P <0.001).This study demonstrates that the bone textural image analysis technique can be used to quantify the radiographic changes in RA hands, based on comparisons of FDs.


Subject(s)
Arthritis, Rheumatoid/pathology , Cancellous Bone/diagnostic imaging , Hand/diagnostic imaging , Image Processing, Computer-Assisted/methods , Adult , Aged , Aged, 80 and over , Female , Fractals , Humans , Male , Middle Aged , Retrospective Studies
17.
Nat Commun ; 7: 13171, 2016 10 27.
Article in English | MEDLINE | ID: mdl-27786175

ABSTRACT

Control of intestinal epithelial stemness is crucial for tissue homeostasis. Disturbances in epithelial function are implicated in inflammatory and neoplastic diseases of the gastrointestinal tract. Here we report that mitochondrial function plays a critical role in maintaining intestinal stemness and homeostasis. Using intestinal epithelial cell (IEC)-specific mouse models, we show that loss of HSP60, a mitochondrial chaperone, activates the mitochondrial unfolded protein response (MT-UPR) and results in mitochondrial dysfunction. HSP60-deficient crypts display loss of stemness and cell proliferation, accompanied by epithelial release of WNT10A and RSPO1. Sporadic failure of Cre-mediated Hsp60 deletion gives rise to hyperproliferative crypt foci originating from OLFM4+ stem cells. These effects are independent of the MT-UPR-associated transcription factor CHOP. In conclusion, compensatory hyperproliferation of HSP60+ escaper stem cells suggests paracrine release of WNT-related factors from HSP60-deficient, functionally impaired IEC to be pivotal in the control of the proliferative capacity of the stem cell niche.


Subject(s)
Cell Proliferation , Embryonic Stem Cells/metabolism , Intestinal Mucosa/metabolism , Mitochondria/metabolism , Animals , Chaperonin 60/genetics , Chaperonin 60/metabolism , Embryonic Stem Cells/cytology , Female , Gene Expression Regulation, Developmental , Intestinal Mucosa/cytology , Intestinal Mucosa/embryology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Mice, Transgenic , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Unfolded Protein Response/genetics , Wnt Proteins/genetics , Wnt Proteins/metabolism
18.
Front Immunol ; 7: 154, 2016.
Article in English | MEDLINE | ID: mdl-27148273

ABSTRACT

Chronic diseases, such as obesity and diabetes, cardiovascular, and inflammatory bowel diseases (IBD) share common features in their pathology. Metabolic disorders exhibit strong inflammatory underpinnings and vice versa, inflammation is associated with metabolic alterations. Next to cytokines and cellular stress pathways, such as the unfolded protein response (UPR), alterations in the enteroendocrine system are intersections of various pathologies. Enteroendocrine cells (EEC) have been studied extensively for their ability to regulate gastrointestinal motility, secretion, and insulin release by release of peptide hormones. In particular, the L-cell-derived incretin hormone glucagon-like peptide 1 (GLP-1) has gained enormous attention due to its insulinotropic action and relevance in the treatment of type 2 diabetes (T2D). Yet, accumulating data indicate a critical role for EEC and in particular for GLP-1 in metabolic adaptation and in orchestrating immune responses beyond blood glucose control. EEC sense the lamina propria and luminal environment, including the microbiota via receptors and transporters. Subsequently, mediating signals by secreting hormones and cytokines, EEC can be considered as integrators of metabolic and inflammatory signaling. This review focuses on L cell and GLP-1 functions in the context of metabolic and inflammatory diseases. The effects of incretin-based therapies on metabolism and immune system are discussed and the interrelation and common features of metabolic and immune-mediated disorders are highlighted. Moreover, it presents data on the impact of inflammation, in particular of IBD on EEC and discusses the potential role of the microbiota as link between nutrients, metabolism, immunity, and disease.

19.
Sci Rep ; 5: 16831, 2015 Nov 19.
Article in English | MEDLINE | ID: mdl-26582215

ABSTRACT

Intestinal nutrient transport and sensing are of emerging interest in research on obesity and diabetes and as drug targets. Appropriate in vitro models are lacking that allow both, studies on transport processes as well as sensing and subsequent incretin hormone secretion including intracellular signaling. We here demonstrate that murine small-intestinal organoids are the first in vitro model system enabling concurrent investigations of nutrient and drug transport, sensing and incretin hormone secretion as well as fluorescent live-cell imaging of intracellular signaling processes. By generating organoid cultures from wild type mice and animals lacking different nutrient transporters, we show that organoids preserve the main phenotypic features and functional characteristics of the intestine. This turns them into the best in vitro model currently available and opens new avenues for basic as well as medical research.


Subject(s)
Food , Incretins/metabolism , Intestinal Mucosa/metabolism , Organoids/metabolism , Acids/metabolism , Animals , Biological Transport , Calcium/metabolism , Cell Survival , Fluoresceins/metabolism , Fluorescence , Fura-2/analogs & derivatives , Fura-2/metabolism , Intestine, Small/metabolism , Intracellular Space/metabolism , Mice, Inbred C57BL , Molecular Imaging , Pharmaceutical Preparations/metabolism , Reproducibility of Results
20.
Wien Klin Wochenschr ; 127(19-20): 792-4, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26142172

ABSTRACT

Mixed connective tissue disease (MCTD) is a rare connective tissue disease frequently involving the lungs. The main characteristic is a systemic sclerosis-like picture of slowly progressing interstitial lung disease consistent with lung fibrosis, while pulmonary arterial hypertension is rare. Herein, we present a case of a newly diagnosed MCTD patient developing life-threatening acute pneumonitis similar to lupus pneumonitis. Previous literature on this exceptionally rare complication of MCTD is reviewed and differential diagnosis and management discussed.


Subject(s)
Mixed Connective Tissue Disease/diagnosis , Mixed Connective Tissue Disease/therapy , Pneumonia/diagnosis , Pneumonia/therapy , Respiratory Insufficiency/prevention & control , Acute Disease , Adult , Critical Care/methods , Diagnosis, Differential , Female , Humans , Mixed Connective Tissue Disease/complications , Pneumonia/etiology , Respiratory Insufficiency/diagnosis , Respiratory Insufficiency/etiology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...