1
|
Gawlik-Kotelnicka O, Czarnecka-Chrebelska K, Margulska A, Pikus E, Wasiak J, Skowrońska A, Brzeziańska-Lasota E, Strzelecki D. Associations between intestinal fatty-acid binding protein and clinical and metabolic characteristics of depression. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111170. [PMID: 39393435 DOI: 10.1016/j.pnpbp.2024.111170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 10/13/2024]
Abstract
INTRODUCTION The topic of increased intestinal permeability is associated with disruption of the intestinal barrier, leading to the "leaky gut" syndrome. Depressive disorders often coexist with abdominal obesity, metabolic syndrome, or its components and complications. Intestinal permeability has been proven to relate to all of the above. METHODS In this cross-sectional study, we aimed to assess the "leaky gut" blood biomarker - intestinal fatty acid-binding protein (I-FABP) - in 114 adult patients diagnosed with depressive disorders depending on abdominal obesity comorbidity, depression, anxiety, and stress level, or antidepressant use. The corrected p-value was set at 0.02. We analyzed patients' mental state, diet, anthropometric parameters, metabolic laboratory markers and I-FABP. RESULTS There was no difference in circulating I-FABP levels between obese and non-obese patients with depressive disorders (p = 0.648). Similarly, I-FABP levels were not different in patients with different emotional symptoms severity (p = 0.829 for self-assessed depression, p = 0.164 for anxiety, and p = 0.543 for stress). But, I-FABP levels differed significantly between patients treated and not treated with antidepressants (p = 0.011). In general linear model analysis treatment with antidepressants, anxiety severity level, their interaction, along with smoking status, drinks intake, and using dietary supplements were shown to significantly explain I-FABP variance (p < 0.001, R2adj = 0.261). CONCLUSIONS Comorbid obesity did not increase intestinal permeability circulating marker, I-FABP, in the population of patients with depressive disorders. Treatment with antidepressants may be connected to higher I-FABP levels. Using dietary supplements, drinks intake, smoking status, or anxiety level may serve as explanatory factors.
Collapse
Affiliation(s)
- Oliwia Gawlik-Kotelnicka
- Department of Affective and Psychotic Disorders, Medical University of Lodz, Czechosłowacka 8/10, 92-216 Lodz, Poland.
| | | | - Aleksandra Margulska
- Department of Child and Adolescent Psychiatry, Medical University of Lodz, Czechosłowacka 8/10, 92-216 Lodz, Poland.
| | - Ewa Pikus
- Department of Biomedicine and Genetics, Medical University of Lodz, 92-213 Lodz, Poland.
| | - Jakub Wasiak
- Faculty of Medicine, Medical University of Lodz, Kościuszki 4, 90-419 Lodz, Poland.
| | - Anna Skowrońska
- Department of Affective and Psychotic Disorders, Medical University of Lodz, Czechosłowacka 8/10, 92-216 Lodz, Poland
| | - Ewa Brzeziańska-Lasota
- Department of Biomedicine and Genetics, Medical University of Lodz, 92-213 Lodz, Poland.
| | - Dominik Strzelecki
- Department of Affective and Psychotic Disorders, Medical University of Lodz, Czechosłowacka 8/10, 92-216 Lodz, Poland.
| |
Collapse
|
2
|
Byrne A, Diener C, Brown BP, Maust BS, Feng C, Alinde BL, Gibbons SM, Koch M, Gray CM, Jaspan HB, Nyangahu DD. Neonates exposed to HIV but uninfected exhibit an altered gut microbiota and inflammation associated with impaired breast milk antibody function. MICROBIOME 2024; 12:261. [PMID: 39707483 DOI: 10.1186/s40168-024-01973-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/11/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND Infants exposed to HIV but uninfected have altered immune profiles which include heightened systemic inflammation. The mechanism(s) underlying this phenomenon is unknown. Here, we investigated differences in neonatal gut bacterial and viral microbiome and associations with inflammatory biomarkers in plasma. Further, we tested whether HIV exposure impacts antibody-microbiota binding in neonatal gut and whether antibodies in breast milk impact the growth of commensal bacteria. RESULTS Neonates exposed to HIV but uninfected (nHEU) exhibited altered gut bacteriome and virome compared to unexposed neonates (nHU). In addition, HIV exposure differentially impacted IgA-microbiota binding in neonates. The relative abundance of Blautia spp. in the whole stool or IgA-bound microbiota was positively associated with plasma concentrations of C-reactive protein. Finally, IgA from the breast milk of mothers living with HIV displayed a significantly lower ability to inhibit the growth of Blautia coccoides which was associated with inflammation in nHEU. CONCLUSION nHEU exhibits profound alterations in gut bacterial microbiota with a mild impact on the enteric DNA virome. Elevated inflammation in nHEU could be due to a lower capacity of breast milk IgA from mothers living with HIV to limit growth the of gut bacteria associated with inflammation. Video Abstract.
Collapse
Affiliation(s)
- Audrey Byrne
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Christian Diener
- Institute For Systems Biology, Seattle, WA, 98109, USA
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Graz, Austria
| | - Bryan P Brown
- Seattle Children's Research Institute, Seattle, WA, USA
| | | | - Colin Feng
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Berenice L Alinde
- Stellenbosch University, Cape Town, South Africa
- Division of Immunology, University of Cape Town, Cape Town, South Africa
| | - Sean M Gibbons
- Institute For Systems Biology, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA
- eScience Institute, University of Washington, Seattle, WA, 98195, USA
| | - Meghan Koch
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98101, USA
- Department of Immunology, University of Washington, Seattle, WA, 98195, USA
| | - Clive M Gray
- Stellenbosch University, Cape Town, South Africa
- Division of Immunology, University of Cape Town, Cape Town, South Africa
| | - Heather B Jaspan
- Seattle Children's Research Institute, Seattle, WA, USA
- Division of Immunology, University of Cape Town, Cape Town, South Africa
- Department of Pediatrics, University of Washington, Seattle, WA, 98195, USA
| | - Donald D Nyangahu
- Seattle Children's Research Institute, Seattle, WA, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, 98195, USA.
- Department of Pharmacology, The State University of New Jersey, RutgersPiscataway, NJ, 08854, USA.
- Center for Advanced Biotechnology and Medicine, The State University of New Jersey, RutgersPiscataway, NJ, 08854, USA.
- Department of Human Pathology, Egerton University, Nakuru, Kenya.
| |
Collapse
|
3
|
Zhang Q, Zhao L, Li Y, Wang S, Lu G, Wang H. Advances in the mechanism of action of short-chain fatty acids in psoriasis. Int Immunopharmacol 2024; 141:112928. [PMID: 39159566 DOI: 10.1016/j.intimp.2024.112928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/22/2024] [Accepted: 08/10/2024] [Indexed: 08/21/2024]
Abstract
Psoriasis is a prevalent chronic inflammatory and immunological disorder. Its lesions are present as scaly erythema or plaques. Disruptions in the body's immune system play a significant role in developing psoriasis. Recent evidence suggests a potential role of the gut microbiome in autoimmune diseases. Short-chain fatty acids (SCFAs) are the primary metabolites created by gut microbes and play a crucial fuction in autoimmunity. SCFAs act on various cells by mediating signaling to participate in host physiological and pathological processes. These processes encompass body metabolism, maintenance of intestinal barrier function, and immune system modulation. SCFAs can regulate immune cells to enhance the body's immune function, potentially influencing the prevention and treatment of psoriasis. However, the mechanisms underlying the role of SCFAs in psoriasis remain incompletely understood. This paper examines the relationship between SCFAs and psoriasis, elucidating how SCFAs influence the immune system, inflammatory response, and gut barrier in psoriasis. According to the study, in psoriasis, SCFAs have been shown to regulate neutrophils, macrophages, and dendritic cells in the adaptive immune system, as well as T and B cells in the innate immune system. Additionally, we explore the role of SCFAs in psoriasis by maintaining intestinal barrier function, restoring intestinal ecological homeostasis, and investigating the potential therapeutic benefits of SCFAs for psoriasis.
Collapse
Affiliation(s)
- Qin Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China; Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Linna Zhao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China; Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China.
| | - Yu Li
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China; Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Siyao Wang
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China; Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guiling Lu
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
| | - Hongmei Wang
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China.
| |
Collapse
|
4
|
Xi M, Ruan Q, Zhong S, Li J, Qi W, Xie C, Wang X, Abuduxiku N, Ni J. Periodontal bacteria influence systemic diseases through the gut microbiota. Front Cell Infect Microbiol 2024; 14:1478362. [PMID: 39619660 PMCID: PMC11604649 DOI: 10.3389/fcimb.2024.1478362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/29/2024] [Indexed: 12/11/2024] Open
Abstract
Many systemic diseases, including Alzheimer disease (AD), diabetes mellitus (DM) and cardiovascular disease, are associated with microbiota dysbiosis. The oral and intestinal microbiota are directly connected anatomically, and communicate with each other through the oral-gut microbiome axis to establish and maintain host microbial homeostasis. In addition to directly, periodontal bacteria may also be indirectly involved in the regulation of systemic health and disease through the disturbed gut. This paper provides evidence for the role of periodontal bacteria in systemic diseases via the oral-gut axis and the far-reaching implications of maintaining periodontal health in reducing the risk of many intestinal and parenteral diseases. This may provide insight into the underlying pathogenesis of many systemic diseases and the search for new preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Mengying Xi
- Department of Periodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Qijun Ruan
- Department of Periodontics, Shenzhen Longgang Otolaryngology hospital, Shenzhen, China
| | - Sulan Zhong
- Department of Periodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Jiatong Li
- Department of Periodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Weijuan Qi
- Department of Periodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Congman Xie
- Department of Orthodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Xiaoyan Wang
- Department of Periodontics, Shenzhen Longgang Otolaryngology hospital, Shenzhen, China
| | - Nuerbiya Abuduxiku
- Department of Stomatology, The First People’s Hospital of Kashi, Kashi, China
| | - Jia Ni
- Department of Periodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| |
Collapse
|
5
|
Miuma S, Miyaaki H, Taura N, Kanda Y, Matsuo S, Tajima K, Takahashi K, Nakao Y, Fukushima M, Haraguchi M, Sasaki R, Ozawa E, Ichikawa T, Nakao K. Elevated intestinal fatty acid-binding protein levels as a marker of portal hypertension and gastroesophageal varices in cirrhosis. Sci Rep 2024; 14:25003. [PMID: 39443545 PMCID: PMC11499902 DOI: 10.1038/s41598-024-76040-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024] Open
Abstract
We measured intestinal fatty acid-binding protein (I-FABP) levels, a useful marker of small intestinal mucosal injury, in patients with cirrhosis to determine their relationship with liver function and complications. This cross-sectional study included 71 patients with cirrhosis admitted for treatment of cirrhotic complications or hepatocellular carcinoma (cohort A) and 104 patients with cirrhosis who received direct-acting antiviral therapy for HCV (cohort B). I-FABP levels, measured by ELISA, were evaluated relative to hepatic reserve and compared with non-invasive scoring systems for diagnostic performance in cirrhotic complications. The median I-FABP level in both cohorts were significantly elevated in patients with reduced hepatic reserve (CTP grade A/BC cohort A, 2.33/3.17 ng/mL, p = 0.032; cohort B, 2.46/3.64 ng/mL, p = 0.008) and complications with gastroesophageal varices (GEV; GEV (-)/(+) cohort A, 1.66/3.67 ng/mL, p < 0.001; cohort B, 2.32/3.36 ng/mL; p = 0.003). Further, multiple logistic regression analysis identified I-FABP as the only factor contributing to GEV presence in both cohorts, which outperformed non-invasive scoring systems for GEV diagnosis (sensitivity 84.6%; specificity 84.2%; sensitivity 69.6%; specificity 63.8%, respectively). In conclusion, elevated small-intestinal mucosal injury in patients with cirrhosis was related to reduced hepatic reserve and GEV presence. I-FABP levels reflect portal hypertension and may be useful in cirrhosis management.
Collapse
Affiliation(s)
- Satoshi Miuma
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, 1- 7-1 Sakamoto, Nagasaki, 852-8501, Japan.
| | - Hisamitsu Miyaaki
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, 1- 7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Naota Taura
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, 1- 7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Yasuko Kanda
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, 1- 7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Satoshi Matsuo
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, 1- 7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Kazuaki Tajima
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, 1- 7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Kosuke Takahashi
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, 1- 7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Yasuhiko Nakao
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, 1- 7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Masanori Fukushima
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, 1- 7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Masafumi Haraguchi
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, 1- 7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Ryu Sasaki
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, 1- 7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Eisuke Ozawa
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, 1- 7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Tatsuki Ichikawa
- Department of Gastroenterology, Nagasaki Harbor Medical Center, Shinti 6-39, Nagasaki, 850-8555, Japan
| | - Kazuhiko Nakao
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, 1- 7-1 Sakamoto, Nagasaki, 852-8501, Japan
| |
Collapse
|
6
|
Radaschin DS, Tatu A, Iancu AV, Beiu C, Popa LG. The Contribution of the Skin Microbiome to Psoriasis Pathogenesis and Its Implications for Therapeutic Strategies. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1619. [PMID: 39459406 PMCID: PMC11509136 DOI: 10.3390/medicina60101619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 09/26/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024]
Abstract
Psoriasis is a common chronic inflammatory skin disease, associated with significant morbidity and a considerable negative impact on the patients' quality of life. The complex pathogenesis of psoriasis is still incompletely understood. Genetic predisposition, environmental factors like smoking, alcohol consumption, psychological stress, consumption of certain drugs, and mechanical trauma, as well as specific immune dysfunctions, contribute to the onset of the disease. Mounting evidence indicate that skin dysbiosis plays a significant role in the development and exacerbation of psoriasis through loss of immune tolerance to commensal skin flora, an altered balance between Tregs and effector cells, and an excessive Th1 and Th17 polarization. While the implications of skin dysbiosis in psoriasis pathogenesis are only starting to be revealed, the progress in the characterization of the skin microbiome changes in psoriasis patients has opened a whole new avenue of research focusing on the modulation of the skin microbiome as an adjuvant treatment for psoriasis and as part of a long-term plan to prevent disease flares. The skin microbiome may also represent a valuable predictive marker of treatment response and may aid in the selection of the optimal personalized treatment. We present the current knowledge on the skin microbiome changes in psoriasis and the results of the studies that investigated the efficacy of the different skin microbiome modulation strategies in the management of psoriasis, and discuss the complex interaction between the host and skin commensal flora.
Collapse
Affiliation(s)
- Diana Sabina Radaschin
- Department of Clinical Medical, Faculty of Medicine and Pharmacy, “Saint Parascheva” Infectious Disease Clinical Hospital, Multidisciplinary Integrated Centre of Dermatological Interface Research Centre (MICDIR), “Dunarea de Jos” University of Galati, 800008 Galati, Romania
| | - Alin Tatu
- Department of Clinical Medical, Faculty of Medicine and Pharmacy, “Saint Parascheva” Infectious Disease Clinical Hospital, Multidisciplinary Integrated Centre of Dermatological Interface Research Centre (MICDIR), “Dunarea de Jos” University of Galati, 800008 Galati, Romania
| | - Alina Viorica Iancu
- Department of Morphological and Functional Sciences, “Dunarea de Jos” University of Galati, 800008 Galati, Romania
| | - Cristina Beiu
- Department of Oncologic Dermatology, Elias Emergency University Hospital, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Liliana Gabriela Popa
- Department of Oncologic Dermatology, Elias Emergency University Hospital, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
7
|
Dmytriv TR, Storey KB, Lushchak VI. Intestinal barrier permeability: the influence of gut microbiota, nutrition, and exercise. Front Physiol 2024; 15:1380713. [PMID: 39040079 PMCID: PMC11260943 DOI: 10.3389/fphys.2024.1380713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 05/29/2024] [Indexed: 07/24/2024] Open
Abstract
The intestinal wall is a selectively permeable barrier between the content of the intestinal lumen and the internal environment of the body. Disturbances of intestinal wall permeability can potentially lead to unwanted activation of the enteric immune system due to excessive contact with gut microbiota and its components, and the development of endotoxemia, when the level of bacterial lipopolysaccharides increases in the blood, causing chronic low-intensity inflammation. In this review, the following aspects are covered: the structure of the intestinal wall barrier; the influence of the gut microbiota on the permeability of the intestinal wall via the regulation of functioning of tight junction proteins, synthesis/degradation of mucus and antioxidant effects; the molecular mechanisms of activation of the pro-inflammatory response caused by bacterial invasion through the TLR4-induced TIRAP/MyD88 and TRAM/TRIF signaling cascades; the influence of nutrition on intestinal permeability, and the influence of exercise with an emphasis on exercise-induced heat stress and hypoxia. Overall, this review provides some insight into how to prevent excessive intestinal barrier permeability and the associated inflammatory processes involved in many if not most pathologies. Some diets and physical exercise are supposed to be non-pharmacological approaches to maintain the integrity of intestinal barrier function and provide its efficient operation. However, at an early age, the increased intestinal permeability has a hormetic effect and contributes to the development of the immune system.
Collapse
Affiliation(s)
- Tetiana R. Dmytriv
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
- Research and Development University, Ivano-Frankivsk, Ukraine
| | | | - Volodymyr I. Lushchak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
- Research and Development University, Ivano-Frankivsk, Ukraine
| |
Collapse
|
8
|
Liu YC, Chuang SH, Chen YP, Shih YH. Associations of novel complete blood count-derived inflammatory markers with psoriasis: a systematic review and meta-analysis. Arch Dermatol Res 2024; 316:228. [PMID: 38787437 DOI: 10.1007/s00403-024-02994-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 04/15/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024]
Abstract
Psoriasis is an immune-mediated disorder which primarily affects skin and has systemic inflammatory involvement. Neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), systemic immune-inflammation index (SII), and monocyte-to-lymphocyte ratio (MLR) are novel complete blood count (CBC)-derived markers which can reflect systemic inflammation. This study aimed to systematically investigate the associations of NLR, PLR, SII, and MLR with psoriasis. This study was performed in compliance with the Preferred Reporting Items for Systematic Reviews and Meta-analyses statement. A comprehensive search of Pubmed, Embase, Scopus, and Google Scholar was conducted for relevant studies. Observational studies evaluating the correlations of NLR, PLR, SII, or MLR with psoriasis were included. The primary outcomes were the associations of these inflammatory markers with the presence and severity of psoriasis. The random-effect model was applied for meta-analysis. 36 studies comprising 4794 psoriasis patients and 55,121 individuals in total were included in the meta-analysis. All inflammatory markers were significantly increased in psoriasis groups compared to healthy controls (NLR: MD = 0.59, 95% CI: 0.47-0.7; PLR: MD = 15.53, 95% CI: 8.48-22.58; SII: MD = 111.58, 95% CI: 61.49-161.68; MLR: MD = 0.034, 95% CI: 0.021-0.048; all p < 0.001). Between-group mean differences in NLR and PLR were positively correlated with the mean scores of Psoriasis Area Severity Index (NLR: p = 0.041; PLR: p = 0.021). NLR, PLR, SII, and MLR are associated with the presence of psoriasis. NLR and PLR serve as significant indicators of psoriasis severity. These novel CBC-derived markers constitute potential targets in the screening and monitoring of psoriasis.
Collapse
Affiliation(s)
- Yu-Cheng Liu
- Department of General Medicine, Taipei Medical University Shuang Ho Hospital, New Taipei, 23561, Taiwan
| | - Shu-Han Chuang
- Division of General Practice, Department of Medical Education, Changhua Christian Hospital, Changhua, 50006, Taiwan
| | - Yu-Pin Chen
- Department of Orthopedics, Taipei Municipal Wan Fang Hospital, Taipei, 11696, Taiwan
- Department of Orthopedics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Yi-Hsien Shih
- Department of Dermatology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
- Department of Dermatology, Taipei Medical University Shuang Ho Hospital, New Taipei City, 23561, Taiwan.
| |
Collapse
|
9
|
Pohjonen J, Kaukinen K, Huhtala H, Pörsti I, Lindfors K, Mustonen J, Mäkelä S. Indirect Markers of Intestinal Damage in IgA Nephropathy. Nephron Clin Pract 2024; 148:693-700. [PMID: 38723612 PMCID: PMC11460831 DOI: 10.1159/000538242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 03/05/2024] [Indexed: 06/11/2024] Open
Abstract
INTRODUCTION Presence of subclinical intestinal inflammation has repeatedly been shown in IgA nephropathy (IgAN) and the degree of histological inflammation has correlated with abnormal urinary findings. There is lack of noninvasive biomarkers evaluating the presence of subclinical intestinal damage in IgAN. We conducted this study hypothesizing that selected biomarkers regarded as indirect markers of intestinal damage could be elevated in IgAN. METHODS Eighty-five primary IgAN patients (median age 55 years, 54% men) participated in this single-center study in Tampere, Finland. None had end-stage kidney disease or previously diagnosed enteropathies. Celiac disease was excluded with serum transglutaminase 2 antibody (TG2Ab) and endomysial antibody tests and inflammatory bowel disease with fecal calprotectin. Intestinal damage was evaluated from sera with analyses of intestinal fatty-acid binding protein (I-FABP), soluble cluster of differentiation molecule 14 (sCD14), and lipopolysaccharide binding protein. Fourteen people suffering from dyspepsia and 15 healthy people served as controls. RESULTS I-FABP levels among IgAN patients were higher than in the healthy controls (median 830 pg/mL vs. 289 pg/mL, p < 0.001). Also, sCD14 was increased in IgAN patients compared to dyspepsia controls. Although TG2Ab levels were within the normal range among IgAN patients, they were higher than in the healthy controls (median 1.3 U/mL vs. 0.6 U/mL, p < 0.001). CONCLUSIONS Elevated serum levels of I-FABP were present in primary IgAN patients without known enteropathies. Serum I-FABP may indicate the presence of subclinical intestinal damage. These findings encourage further investigation into the role of the intestine in the pathophysiology of IgAN.
Collapse
Affiliation(s)
- Jussi Pohjonen
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Internal Medicine, Tampere University Hospital, Tampere, Finland
| | - Katri Kaukinen
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Internal Medicine, Tampere University Hospital, Tampere, Finland
| | - Heini Huhtala
- Faculty of Social Sciences, Tampere University, Tampere, Finland
| | - Ilkka Pörsti
- Department of Internal Medicine, Tampere University Hospital, Tampere, Finland
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Katri Lindfors
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Jukka Mustonen
- Department of Internal Medicine, Tampere University Hospital, Tampere, Finland
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Satu Mäkelä
- Department of Internal Medicine, Tampere University Hospital, Tampere, Finland
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
10
|
Hernandez-Nicols BF, Robledo-Pulido JJ, Alvarado-Navarro A. Etiopathogenesis of Psoriasis: Integration of Proposed Theories. Immunol Invest 2024; 53:348-415. [PMID: 38240030 DOI: 10.1080/08820139.2024.2302823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2024]
Abstract
Psoriasis is a chronic inflammatory disease characterized by squamous and erythematous plaques on the skin and the involvement of the immune system. Global prevalence for psoriasis has been reported around 1-3% with a higher incidence in adults and similar proportions between men and women. The risk factors associated with psoriasis are both extrinsic and intrinsic, out of which a polygenic predisposition is a highlight out of the latter. Psoriasis etiology is not yet fully described, but several hypothesis have been proposed: 1) the autoimmunity hypothesis is based on the over-expression of antimicrobial peptides such as LL-37, the proteins ADAMTSL5, K17, and hsp27, or lipids synthesized by the PLA2G4D enzyme, all of which may serve as autoantigens to promote the differentiation of autoreactive lymphocytes T and unleash a chronic inflammatory response; 2) dysbiosis of skin microbiota hypothesis in psoriasis has gained relevance due to the observations of a loss of diversity and the participation of pathogenic bacteria such as Streptococcus spp. or Staphylococcus spp. the fungi Malassezia spp. or Candida spp. and the virus HPV, HCV, or HIV in psoriatic plaques; 3) the oxidative stress hypothesis, the most recent one, describes that the cell injury and the release of proinflammatory mediators and antimicrobial peptides that leads to activate of the Th1/Th17 axis observed in psoriasis is caused by a higher release of reactive oxygen species and the imbalance between oxidant and antioxidant mechanisms. This review aims to describe the mechanisms involved in the three hypotheses on the etiopathogeneses of psoriasis.
Collapse
Affiliation(s)
- Brenda Fernanda Hernandez-Nicols
- Centro de Investigación en Inmunología y Dermatología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Juan José Robledo-Pulido
- Centro de Investigación en Inmunología y Dermatología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Anabell Alvarado-Navarro
- Centro de Investigación en Inmunología y Dermatología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| |
Collapse
|
11
|
Liu S, He M, Jiang J, Duan X, Chai B, Zhang J, Tao Q, Chen H. Triggers for the onset and recurrence of psoriasis: a review and update. Cell Commun Signal 2024; 22:108. [PMID: 38347543 PMCID: PMC10860266 DOI: 10.1186/s12964-023-01381-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 11/02/2023] [Indexed: 02/15/2024] Open
Abstract
Psoriasis is an immune-mediated inflammatory skin disease, involving a complex interplay between genetic and environmental factors. Previous studies have demonstrated that genetic factors play a major role in the pathogenesis of psoriasis. However, non-genetic factors are also necessary to trigger the onset and recurrence of psoriasis in genetically predisposed individuals, which include infections, microbiota dysbiosis of the skin and gut, dysregulated lipid metabolism, dysregulated sex hormones, and mental illness. Psoriasis can also be induced by other environmental triggers, such as skin trauma, unhealthy lifestyles, and medications. Understanding how these triggers play a role in the onset and recurrence of psoriasis provides insights into psoriasis pathogenesis, as well as better clinical administration. In this review, we summarize the triggers for the onset and recurrence of psoriasis and update the current evidence on the underlying mechanism of how these factors elicit the disease. Video Abstract.
Collapse
Grants
- No.82173423, No.81974475, No.82103731 the National Natural Science Foundation of China
- No.82173423, No.81974475, No.82103731 the National Natural Science Foundation of China
- No.82173423, No.81974475, No.82103731 the National Natural Science Foundation of China
- Basic Research Project, No. JCYJ20190809103805589 Shenzhen Natural Science Foundation
- Basic Research Project, No. JCYJ20190809103805589 Shenzhen Natural Science Foundation
- Basic Research Project, No. JCYJ20190809103805589 Shenzhen Natural Science Foundation
- Key Project, No.2019003 Shenzhen Nanshan District Science and Technology Project
- Key Project, No.2019003 Shenzhen Nanshan District Science and Technology Project
- Key Project, No.2019003 Shenzhen Nanshan District Science and Technology Project
Collapse
Affiliation(s)
- Suwen Liu
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Mengwen He
- Department of Dermatology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China
| | - Jian Jiang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaoru Duan
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bao Chai
- Department of Dermatology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China
- Department of Dermatology, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, 518052, China
| | - Jingyu Zhang
- Department of Dermatology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China
- Department of Dermatology, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, 518052, China
| | - Qingxiao Tao
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hongxiang Chen
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Department of Dermatology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China.
| |
Collapse
|
12
|
Tie Y, Huang Y, Chen R, Li L, Chen M, Zhang S. Current insights on the roles of gut microbiota in inflammatory bowel disease-associated extra-intestinal manifestations: pathophysiology and therapeutic targets. Gut Microbes 2023; 15:2265028. [PMID: 37822139 PMCID: PMC10572083 DOI: 10.1080/19490976.2023.2265028] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 09/26/2023] [Indexed: 10/13/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic, recurrent inflammatory disease of the gastrointestinal tract. In addition to digestive symptoms, patients with IBD may also develop extra-intestinal manifestations (EIMs), the etiology of which remains undefined. The gut microbiota has been reported to exert a critical role in the pathogenesis of IBD, with a similar pattern of gut dysbiosis observed between patients with IBD and those with EIMs. Therefore, it is hypothesized that the gut microbiota is also involved in the pathogenesis of EIMs. The potential mechanisms are presented in this review, including: 1) impaired gut barrier: dysbiosis induces pore formation in the intestinal epithelium, and activates pattern recognition receptors to promote local inflammation; 2) microbial translocation: intestinal pathogens, antigens, and toxins translocate via the impaired gut barrier into extra-intestinal sites; 3) molecular mimicry: certain microbial antigens share similar epitopes with self-antigens, inducing inflammatory responses targeting extra-intestinal tissues; 4) microbiota-related metabolites: dysbiosis results in the dysregulation of microbiota-related metabolites, which could modulate the differentiation of lymphocytes and cytokine production; 5) immunocytes and cytokines: immunocytes are over-activated and pro-inflammatory cytokines are excessively released. Additionally, we summarize microbiota-related therapies, including probiotics, prebiotics, postbiotics, antibiotics, and fecal microbiota transplantation, to promote better clinical management of IBD-associated EIMs.
Collapse
Affiliation(s)
- Yizhe Tie
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yongle Huang
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Clinical Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Rirong Chen
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li Li
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Minhu Chen
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shenghong Zhang
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
13
|
Joshi M, Hiremath P, John J, Ranadive N, Nandakumar K, Mudgal J. Modulatory role of vitamins A, B3, C, D, and E on skin health, immunity, microbiome, and diseases. Pharmacol Rep 2023; 75:1096-1114. [PMID: 37673852 PMCID: PMC10539462 DOI: 10.1007/s43440-023-00520-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/17/2023] [Accepted: 08/18/2023] [Indexed: 09/08/2023]
Abstract
Disruption of the skin barrier and immunity has been associated with several skin diseases, namely atopic dermatitis (AD), psoriasis, and acne. Resident and non-resident immune cells and the barrier system of the skin are integral to innate immunity. Recent advances in understanding skin microbiota have opened the scope of further understanding the various communications between these microbiota and skin immune cells. Vitamins, being one of the important micronutrients, have been reported to exert antioxidant, anti-inflammatory, and anti-microbial effects. The immunomodulatory action of vitamins can halt the progression of skin diseases, and thus, understanding the immuno-pharmacology of these vitamins, especially for skin diseases can pave the way for their therapeutic potential. At the same time, molecular and cellular markers modulated with these vitamins and their derivatives need to be explored. The present review is focused on significant vitamins (vitamins A, B3, C, D, and E) consumed as nutritional supplements to discuss the outcomes and scope of studies related to skin immunity, health, and diseases.
Collapse
Affiliation(s)
- Mahika Joshi
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Priyanka Hiremath
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Jeena John
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Niraja Ranadive
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Krishnadas Nandakumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Jayesh Mudgal
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| |
Collapse
|
14
|
Cao VT, Carter MC, Brenchley JM, Bolan H, Scott LM, Bai Y, Metcalfe DD, Komarow HD. sCD14 and Intestinal Fatty Acid Binding Protein Are Elevated in the Serum of Patients With Idiopathic Anaphylaxis. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2023; 11:2080-2086.e5. [PMID: 36997122 PMCID: PMC10411508 DOI: 10.1016/j.jaip.2023.03.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 02/02/2023] [Accepted: 03/14/2023] [Indexed: 03/31/2023]
Abstract
BACKGROUND Intestinal epithelial integrity compromise has been identified in gastrointestinal (GI), atopic, and autoimmune diseases. OBJECTIVE Episodes of idiopathic anaphylaxis (IA) are often accompanied by GI manifestations. We, therefore, sought to determine whether surrogate markers of GI permeability were aberrant in this patient population. METHODS Serum concentrations of zonulin, intestinal fatty acid binding protein (I-FABP), and soluble CD14 (sCD14) measured in 54 patients with IA were compared with concentrations in healthy controls (HCs); and correlated with clinical and laboratory parameters. RESULTS The I-FABP was elevated in sera of patients with IA compared with HCs (median 1,378.0 pg/mL vs 479.0 pg/mL, respectively; P < .001). The sCD14 was also elevated compared with HCs (median 2,017.0 ng/mL and 1,189.0 ng/mL, respectively; P < .001), whereas zonulin was comparable between patients with IA and HCs (median 49.6 ng/mL vs 52.4 ng/mL, respectively; P = .40). The I-FABP was elevated in patients with IA who experienced vomiting and/or diarrhea compared with patients with IA who did not (P = .0091). CONCLUSIONS The I-FABP and sCD14 are elevated in the serum of patients with IA. Elevations in these biomarkers of IA provides evidence that increased GI permeability, as is observed in other allergic conditions such as food allergy, is a common finding in those with IA and offers possible insight into the pathogenesis of this disease.
Collapse
Affiliation(s)
- Vivian T Cao
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Melody C Carter
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Jason M Brenchley
- Barrier Immunity Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Hyejeong Bolan
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Linda M Scott
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Yun Bai
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Dean D Metcalfe
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Hirsh D Komarow
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md.
| |
Collapse
|
15
|
Zaragoza-García O, Gutiérrez-Pérez IA, Briceño O, Villafan-Bernal JR, Navarro-Zarza JE, Parra-Rojas I, Falfán-Valencia R, Guzmán-Guzmán IP. IFABP2 as a new prognostic biomarker for secondary non-response in rheumatoid arthritis. Int Immunopharmacol 2023; 119:110090. [PMID: 37044032 DOI: 10.1016/j.intimp.2023.110090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/09/2023] [Accepted: 03/22/2023] [Indexed: 04/14/2023]
Abstract
BACKGROUND Increased intestinal permeability promotes the translocation of bacterial products from the local microbiome to the circulation, inducing inflammation and increasing clinical activity in rheumatoid arthritis (RA). This study evaluates whether intestinal fatty acid binding protein 2 (IFABP2) serum levels are prognostic biomarkers of non-response to conventional synthetic disease-modifying antirheumatic drug therapy (csDMARDs) in RA. METHODS The therapeutic schemes administered to 60 women with RA for at least 18 months were assessed retrospectively, and the treatment response was classified according to the change in DAS28-ESR over time. Serum levels of IFABP2 and TNF-α were determined by ELISA. Receiver operating characteristics (ROC) curve analysis and logistic regression models were used to assess the predictive value and the association of IFABP2 with the non-responder phenotype in RA patients. RESULTS Eleven women had a responder phenotype, 23 had a primary non-responder phenotype, and 26 had a secondary non-responder phenotype. Secondary non-responders showed higher DAS28-ESR (P = 0.009) and higher IFABP2 serum levels compared to the responder group (P = 0.023) and the primary non-responder group (P = 0.018). IFABP2 serum levels were positively correlated with chloroquine dose (r = 0.581, P = 0.007) and negatively correlated with total cholesterol (r = -0.456, P = 0.019) in secondary non-responders. The area under the curve (AUC) value of IFABP2 for predicting secondary non-response was 0.736, and IFABP2 serum levels > 9.311 ng/mL were associated with secondary non-response to csDMARDs (OR = 6.00, P = 0.003). CONCLUSION IFABP2 serum levels are potentially a new biomarker predictive of secondary non-response to csDMARDs in RA, although our findings should be validated externally and in a larger cohort.
Collapse
Affiliation(s)
- Oscar Zaragoza-García
- Faculty of Chemical-Biological Sciences, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| | | | - Olivia Briceño
- Infectious Diseases Research Center, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - José Rafael Villafan-Bernal
- Laboratory of Immunogenomics and Metabolic Diseases, Instituto Nacional de Medicina Genomica, Mexico City, Mexico
| | - José Eduardo Navarro-Zarza
- Department of Medical Internal. Hospital General Dr. RaymundAbarca Alarcón. Chilpancingo, Guerrero, Mexico
| | - Isela Parra-Rojas
- Faculty of Chemical-Biological Sciences, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| | - Ramcés Falfán-Valencia
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Iris Paola Guzmán-Guzmán
- Faculty of Chemical-Biological Sciences, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico.
| |
Collapse
|
16
|
Paradoxical Reactions to Anti-TNFα and Anti-IL-17 Treatment in Psoriasis Patients: Are Skin and/or Gut Microbiota Involved? Dermatol Ther (Heidelb) 2023; 13:911-933. [PMID: 36929119 DOI: 10.1007/s13555-023-00904-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 02/14/2023] [Indexed: 03/18/2023] Open
Abstract
Psoriasis is a chronic, immune-mediated, inflammatory disease primarily affecting the skin. It is currently coming to light that patients with psoriasis have disrupted intestinal barrier and often suffer from comorbidities associated with the gastrointestinal tract. Moreover, there is growing evidence of both cutaneous and intestinal paradoxical reactions during biologic treatment in patients with psoriasis. This review focuses on barrier defects and changes in immune responses in patients with psoriasis, which play an important role in the development of the disease but are also influenced by modern biological treatments targeting IL-17 and TNFα cytokines. Here, we highlight the relationship between the gut-skin axis, microbiota, psoriasis treatment, and the incidence of paradoxical reactions, such as inflammatory bowel disease in patients with psoriasis. A better understanding of the interconnection of these mechanisms could lead to a more personalized therapy and lower the incidence of treatment side effects, thereby improving the quality of life of the affected patients.
Collapse
|
17
|
Reiss Z, Rob F, Kolar M, Schierova D, Kreisinger J, Jackova Z, Roubalova R, Coufal S, Mihula M, Thon T, Bajer L, Novakova M, Vasatko M, Kostovcikova K, Galanova N, Lukas M, Kverka M, Tresnak Hercogova J, Tlaskalova-Hogenova H, Jiraskova Zakostelska Z. Skin microbiota signature distinguishes IBD patients and reflects skin adverse events during anti-TNF therapy. Front Cell Infect Microbiol 2023; 12:1064537. [PMID: 36704107 PMCID: PMC9872723 DOI: 10.3389/fcimb.2022.1064537] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 12/01/2022] [Indexed: 01/11/2023] Open
Abstract
Crohn's disease (CD) and ulcerative colitis (UC) are two forms of inflammatory bowel disease (IBD), where the role of gut but not skin dysbiosis is well recognized. Inhibitors of TNF have been successful in IBD treatment, but up to a quarter of patients suffer from unpredictable skin adverse events (SkAE). For this purpose, we analyzed temporal dynamics of skin microbiota and serum markers of inflammation and epithelial barrier integrity during anti-TNF therapy and SkAE manifestation in IBD patients. We observed that the skin microbiota signature of IBD patients differs markedly from healthy subjects. In particular, the skin microbiota of CD patients differs significantly from that of UC patients and healthy subjects, mainly in the retroauricular crease. In addition, we showed that anti-TNF-related SkAE are associated with specific shifts in skin microbiota profile and with a decrease in serum levels of L-FABP and I-FABP in IBD patients. For the first time, we showed that shifts in microbial composition in IBD patients are not limited to the gut and that skin microbiota and serum markers of the epithelium barrier may be suitable markers of SkAE during anti-TNF therapy.
Collapse
Affiliation(s)
- Zuzana Reiss
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Filip Rob
- Department of Dermatovenerology, Second Faculty of Medicine, Charles University, University Hospital Bulovka, Prague, Czechia
| | - Martin Kolar
- IBD Clinical and Research Centre ISCARE a.s., Prague, Czechia
| | - Dagmar Schierova
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Jakub Kreisinger
- Department of Zoology, Faculty of Science, Charles University, Prague, Czechia
| | - Zuzana Jackova
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Radka Roubalova
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Stepan Coufal
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Martin Mihula
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Tomas Thon
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Lukas Bajer
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia,Department of Gastroenterology and Hepatology, Institute of Clinical and Experimental Medicine, Prague, Czechia
| | - Michaela Novakova
- Department of Dermatovenerology, Second Faculty of Medicine, Charles University, University Hospital Bulovka, Prague, Czechia
| | - Martin Vasatko
- IBD Clinical and Research Centre ISCARE a.s., Prague, Czechia
| | - Klara Kostovcikova
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Natalie Galanova
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Milan Lukas
- IBD Clinical and Research Centre ISCARE a.s., Prague, Czechia,Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czechia
| | - Miloslav Kverka
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Jana Tresnak Hercogova
- Department of Dermatovenerology, Second Faculty of Medicine, Charles University, University Hospital Bulovka, Prague, Czechia,Prof. Hercogova Dermatology, Prague, Czechia
| | | | - Zuzana Jiraskova Zakostelska
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia,*Correspondence: Zuzana Jiraskova Zakostelska,
| |
Collapse
|
18
|
Gilbert BTP, Lamacchia C, Amend L, Strowig T, Rodriguez E, Palmer G, Finckh A. Brief report: Assessment of mucosal barrier integrity using serological biomarkers in preclinical stages of rheumatoid arthritis. Front Immunol 2023; 14:1117742. [PMID: 36875067 PMCID: PMC9977794 DOI: 10.3389/fimmu.2023.1117742] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
Background The pathogenesis of rheumatoid arthritis (RA) is believed to initiate at mucosal sites. The so-called 'mucosal origin hypothesis of RA' postulates an increased intestinal permeability before disease onset. Several biomarkers, including lipopolysaccharide binding protein (LBP) and intestinal fatty acid binding protein (I-FABP), have been proposed to reflect gut mucosa permeability and integrity, while serum calprotectin is a new inflammation marker proposed in RA. Methods We analyzed serum samples of individuals genetically at increased risk of RA in a nested-case-control study. Participants from a longitudinal cohort of first-degree relatives of RA patients (SCREEN-RA cohort) were divided into three pre-clinical stages of RA, based on the presence of risk factors for subsequent RA onset: 1) low-risk healthy asymptomatic controls; 2) intermediate-risk individuals without symptoms, but with RA-associated auto-immunity; 3) high-risk individuals with clinically suspect arthralgias. Five patients with newly diagnosed RA were also sampled. Serum LBP, I-FABP and calprotectin were measured using commercially available ELISA kits. Results We included 180 individuals genetically at increased risk for RA: 84 asymptomatic controls, 53 individuals with RA-associated autoimmunity and 38 high risk individuals. Serum LBP, I-FAPB or calprotectin concentrations did not differ between individuals in different pre-clinical stages of RA. Conclusion Based on the serum biomarkers LBP, I-FABP and calprotectin, we could not detect any evidence for intestinal injury in pre-clinical stages of RA.
Collapse
Affiliation(s)
- Benoît Thomas P Gilbert
- Division of Rheumatology, Department of Medicine, Geneva University Hospitals, Geneva, Switzerland.,Geneva Centre for Inflammation Research (GCIR), Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Céline Lamacchia
- Division of Rheumatology, Department of Medicine, Geneva University Hospitals, Geneva, Switzerland.,Geneva Centre for Inflammation Research (GCIR), Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Lena Amend
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Till Strowig
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Cluster of Excellence Resolving Infection Susceptibility (RESIST) (EXC 2155), Hannover Medical School, Hannover, Germany.,Center for Individualized Infection Medicine (CiiM), Hannover, Germany
| | - Emiliana Rodriguez
- Division of Rheumatology, Department of Medicine, Geneva University Hospitals, Geneva, Switzerland.,Geneva Centre for Inflammation Research (GCIR), Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Gaby Palmer
- Division of Rheumatology, Department of Medicine, Geneva University Hospitals, Geneva, Switzerland.,Geneva Centre for Inflammation Research (GCIR), Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Axel Finckh
- Division of Rheumatology, Department of Medicine, Geneva University Hospitals, Geneva, Switzerland.,Geneva Centre for Inflammation Research (GCIR), Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
19
|
Ramessur R, Corbett M, Marshall D, Acencio ML, Barbosa IA, Dand N, Di Meglio P, Haddad S, Jensen AH, Koopmann W, Mahil SK, Ostaszewski M, Rahmatulla S, Rastrick J, Saklatvala J, Weidinger S, Wright K, Eyerich K, Ndlovu M, Barker JN, Skov L, Conrad C, Smith CH. Biomarkers of disease progression in people with psoriasis: a scoping review. Br J Dermatol 2022; 187:481-493. [PMID: 35482474 PMCID: PMC9796834 DOI: 10.1111/bjd.21627] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 03/31/2022] [Accepted: 04/26/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND Identification of those at risk of more severe psoriasis and/or associated morbidities offers opportunity for early intervention, reduced disease burden and more cost-effective healthcare. Prognostic biomarkers of disease progression have thus been the focus of intense research, but none are part of routine practice. OBJECTIVES To identify and catalogue candidate biomarkers of disease progression in psoriasis for the translational research community. METHODS A systematic search of CENTRAL, Embase, LILACS and MEDLINE was performed for relevant articles published between 1990 and December 2021. Eligibility criteria were studies involving patients with psoriasis (any age, n ≥ 50) reporting biomarkers associated with disease progression. The main outcomes were any measure of skin severity or any prespecified psoriasis comorbidity. Data were extracted by one reviewer and checked by a second; studies meeting minimal quality criteria (longitudinal design and/or use of methods to control for confounding) were formally assessed for bias. Candidate biomarkers were identified by an expert multistakeholder group using a majority voting consensus exercise, and mapped to relevant cellular and molecular pathways. RESULTS Of 181 included studies, most investigated genomic or proteomic biomarkers associated with disease severity (n = 145) or psoriatic arthritis (n = 30). Methodological and reporting limitations compromised interpretation of findings, most notably a lack of longitudinal studies, and inadequate control for key prognostic factors. The following candidate biomarkers with future potential utility were identified for predicting disease severity: LCE3D, interleukin (IL)23R, IL23A, NFKBIL1 loci, HLA-C*06:02 (genomic), IL-17A, IgG aHDL, GlycA, I-FABP and kallikrein 8 (proteomic), tyramine (metabolomic); psoriatic arthritis: HLA-C*06:02, HLA-B*27, HLA-B*38, HLA-B*08, and variation at the IL23R and IL13 loci (genomic); IL-17A, CXCL10, Mac-2 binding protein, integrin b5, matrix metalloproteinase-3 and macrophage-colony stimulating factor (proteomic) and tyramine and mucic acid (metabolomic); and type 2 diabetes mellitus: variation in IL12B and IL23R loci (genomic). No biomarkers were supported by sufficient evidence for clinical use without further validation. CONCLUSIONS This review provides a comprehensive catalogue of investigated biomarkers of disease progression in psoriasis. Future studies must address the common methodological limitations identified herein to expedite discovery and validation of biomarkers for clinical use. What is already known about this topic? The current treatment paradigm in psoriasis is reactive. There is a need to develop effective risk-stratified management approaches that can proactively attenuate the substantial burden of disease. Prognostic biomarkers of disease progression have therefore been the focus of intense research. What does this study add? This review is the first to scope, collate and catalogue research investigating biomarkers of disease progression in psoriasis. The review identifies potentially promising candidate biomarkers for further investigation and highlights common important limitations that should be considered when designing and conducting future studies in this area.
Collapse
Affiliation(s)
- Ravi Ramessur
- St John’s Institute of Dermatology, School of Basic & Medical Biosciences and Department of Medical & Molecular Genetics, School of Basic & Medical Biosciences, Faculty of Life Sciences & MedicineKing’s College LondonLondonUK
| | - Mark Corbett
- Centre for Reviews and DisseminationUniversity of YorkYorkUK
| | - David Marshall
- Centre for Reviews and DisseminationUniversity of YorkYorkUK
| | - Marcio L. Acencio
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | - Ines A. Barbosa
- St John’s Institute of Dermatology, School of Basic & Medical Biosciences and Department of Medical & Molecular Genetics, School of Basic & Medical Biosciences, Faculty of Life Sciences & MedicineKing’s College LondonLondonUK
| | - Nick Dand
- St John’s Institute of Dermatology, School of Basic & Medical Biosciences and Department of Medical & Molecular Genetics, School of Basic & Medical Biosciences, Faculty of Life Sciences & MedicineKing’s College LondonLondonUK
| | - Paola Di Meglio
- St John’s Institute of Dermatology, School of Basic & Medical Biosciences and Department of Medical & Molecular Genetics, School of Basic & Medical Biosciences, Faculty of Life Sciences & MedicineKing’s College LondonLondonUK
| | | | | | - Witte Koopmann
- Department of Translational MedicineLEO Pharma A/SBallerupDenmark
| | - Satveer K. Mahil
- St John’s Institute of Dermatology, School of Basic & Medical Biosciences and Department of Medical & Molecular Genetics, School of Basic & Medical Biosciences, Faculty of Life Sciences & MedicineKing’s College LondonLondonUK
| | - Marek Ostaszewski
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | | | | | - Jake Saklatvala
- St John’s Institute of Dermatology, School of Basic & Medical Biosciences and Department of Medical & Molecular Genetics, School of Basic & Medical Biosciences, Faculty of Life Sciences & MedicineKing’s College LondonLondonUK
| | - Stephan Weidinger
- Department of Dermatology and AllergyUniversity Hospital Schleswig‐HolsteinKielGermany
| | - Kath Wright
- Centre for Reviews and DisseminationUniversity of YorkYorkUK
| | - Kilian Eyerich
- Department of Dermatology and AllergyTechnical University of MunichMunichGermany
- Division of Dermatology, Department of MedicineKarolinska InsitutetStockholmSweden
| | | | - Jonathan N. Barker
- St John’s Institute of Dermatology, School of Basic & Medical Biosciences and Department of Medical & Molecular Genetics, School of Basic & Medical Biosciences, Faculty of Life Sciences & MedicineKing’s College LondonLondonUK
| | - Lone Skov
- Department of Dermatology and Allergy, Herlev and Gentofte Hospital, Department of Clinical MedicineUniversity of CopenhagenCopenhagenDenmark
| | - Curdin Conrad
- Department of DermatologyLausanne University Hospital CHUV & University of LausanneLausanneSwitzerland
| | - Catherine H Smith
- St John’s Institute of Dermatology, School of Basic & Medical Biosciences and Department of Medical & Molecular Genetics, School of Basic & Medical Biosciences, Faculty of Life Sciences & MedicineKing’s College LondonLondonUK
| | | |
Collapse
|
20
|
Nowowiejska J, Baran A, Flisiak I. Fatty Acid-Binding Proteins in Psoriasis-A Review. Metabolites 2022; 12:metabo12090833. [PMID: 36144237 PMCID: PMC9500650 DOI: 10.3390/metabo12090833] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/29/2022] [Accepted: 08/31/2022] [Indexed: 11/16/2022] Open
Abstract
Psoriasis is one of the most common skin diseases in dermatological practice. It affects about 1–3% of the general population and is associated with different comorbidities, especially metabolic syndrome. Fatty-acid-binding proteins (FABPs) are a family of cytosolic proteins which are an important link in lipid metabolism and transport; moreover, they have different tissue specificity and properties. So far, ten FABPs have been discovered and seven have been investigated in psoriasis. In this review, we discuss the nature of all FABPs and their role in psoriasis. FABPs have different organ and tissue expression, and hence various functions, and may be markers of different disorders. Considering the concentration of a few of them tends to be elevated in psoriasis, it confirms the current perception of psoriasis as a multiorgan disorder associated with plenty of comorbidities. Some FABPs may be also further investigated as biomarkers of psoriasis organ complications. FABP-1 and FABP-5 may become potential markers of metabolic complications and inflammation in psoriasis. FABP-7 could perhaps be further investigated as an indicator of the neurodegenerative processes in psoriatic patients.
Collapse
|
21
|
Gadoin E, Desnues C, d'Orbcastel ER, Bouvier T, Auguet JC, Dagorn L, Moroh JL, Adingra A, Bettarel Y. Fishing for the Microbiome of Tropical Tuna. MICROBIAL ECOLOGY 2022:10.1007/s00248-022-02096-4. [PMID: 35962839 DOI: 10.1007/s00248-022-02096-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 07/29/2022] [Indexed: 06/15/2023]
Abstract
Although tunas represent a significant part of the global fish economy and a major nutritional resource worldwide, their microbiome still remains poorly documented. Here, we conducted an analysis of the taxonomic composition of the bacterial communities inhabiting the gut, skin, and liver of two most consumed tropical tuna species (skipjack and yellowfin), from individuals caught in the Atlantic and Indian oceans. We hypothesized that each organ harbors a specific microbial assemblage whose composition might vary according to different biotic (sex, species) and/or abiotic (environmental) factors. Our results revealed that the composition of the tuna microbiome was totally independent of fish sex, regardless of the species and ocean considered. Instead, the main determinants of observed diversity were (i) tuna species for the gut and (ii) sampling site for the skin mucus layer and (iii) a combination of both parameters for the liver. Interestingly, 4.5% of all amplicon sequence variants (ASV) were shared by the three organs, highlighting the presence of a core-microbiota whose most abundant representatives belonged to the genera Mycoplasma, Cutibacterium, and Photobacterium. Our study also revealed the presence of a unique and diversified bacterial assemblage within the tuna liver, comprising a substantial proportion of potential histamine-producing bacteria, well known for their pathogenicity and their contribution to fish poisoning cases. These results indicate that this organ is an unexplored microbial niche whose role in the health of both the host and consumers remains to be elucidated.
Collapse
Affiliation(s)
- Elsa Gadoin
- MARBEC, Université de Montpellier, CNRS, Ifremer, IRD, Place Eugène Bataillon - Bat 24, 34095, Montpellier, France
| | - Christelle Desnues
- Institut Méditerranéen d'Océanologie (MIO), Aix-Marseille Université, Université de Toulon, CNRS, Campus Technologique Et Scientifique de Luminy, 163 avenue de Luminy - Bat. Méditerranée, 13288, Marseille, IRD, France
| | - Emmanuelle Roque d'Orbcastel
- MARBEC, Université de Montpellier, CNRS, Ifremer, IRD, Place Eugène Bataillon - Bat 24, 34095, Montpellier, France
| | - Thierry Bouvier
- MARBEC, Université de Montpellier, CNRS, Ifremer, IRD, Place Eugène Bataillon - Bat 24, 34095, Montpellier, France
| | - Jean-Christophe Auguet
- MARBEC, Université de Montpellier, CNRS, Ifremer, IRD, Place Eugène Bataillon - Bat 24, 34095, Montpellier, France
| | - Laurent Dagorn
- MARBEC, Université de Montpellier, CNRS, Ifremer, IRD, Place Eugène Bataillon - Bat 24, 34095, Montpellier, France
| | - Jean-Luc Moroh
- Université Peleforo Gbon Coulibaly, Korhogo, Ivory Coast
| | - Antoinette Adingra
- Centre de Recherches Océanologiques (CRO) - 29 rue des pêcheurs, Zone 3, Treichville, BP V 18 00225, Abidjan, Ivory Coast
| | - Yvan Bettarel
- MARBEC, Université de Montpellier, CNRS, Ifremer, IRD, Place Eugène Bataillon - Bat 24, 34095, Montpellier, France.
| |
Collapse
|
22
|
Rungjang A, Meephansan J, Payungporn S, Sawaswong V, Chanchaem P, Pureesrisak P, Wongpiyabovorn J, Thio HB. Alteration of gut microbiota during narrowband ultraviolet B therapy: A preliminary study. Exp Dermatol 2022; 31:1281-1288. [PMID: 35737868 DOI: 10.1111/exd.14631] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/06/2022] [Accepted: 06/20/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Gut microbiome dysbiosis is associated with psoriasis development. A relationship between gut microbiota and psoriasis treatment response has been reported. No study has reported the effect of narrowband ultraviolet B (NBUVB) therapy, a standard treatment of psoriasis, on gut microbiota. QUESTION ADDRESSED This study aimed to evaluate gut microbiota change during NBUVB therapy. EXPERIMENTAL DESIGN Stool samples from 22 participants, including 13 patients with chronic plaque psoriasis and nine healthy controls, were recruited. Fecal microbiota composition was analyzed using 16S rRNA sequencing before and after NBUVB therapy. Serum 25-OH Vitamin D of patients with psoriasis was evaluated simultaneously. RESULTS The most abundant phyla of gut microbiota in patients with psoriasis were Firmicutes, Bacteroidetes, Proteobacteria, and Actinobacteria in all participants. Bilophila, Paraprevotella, Alistipes, Sutterella, Romboutsia, Clostridium sensu stricto, and Agathobacter are significantly more enriched in healthy controls. Lactobacillales and Ruminococus torques appeared more enriched after NBUVB treatment in responders but not non-responders. Serum Vitamin D levels significantly increased after NBUVB treatment. CONCLUSIONS&PERSPECTIVE The present study revealed that gut microbiota altered after NBUVB treatment. The change might be treatment-specific and influence the treatment response.
Collapse
Affiliation(s)
- A Rungjang
- Division of Dermatology, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, Thailand
| | - J Meephansan
- Division of Dermatology, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, Thailand
| | - S Payungporn
- The Research Unit of Systems Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - V Sawaswong
- The Research Unit of Systems Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - P Chanchaem
- The Research Unit of Systems Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - P Pureesrisak
- Division of Dermatology, Department of Medicine, Rajavithi Hospital, Ministry of Public Health, Bangkok, Thailand
| | - J Wongpiyabovorn
- Division of Immunology, Department of Microbiology, Faculty of Medicine, Center of Excellence in Immunology and Immune-Mediated Diseases, Chulalongkorn University, Bangkok, Thailand
| | - H B Thio
- Department of Dermatology, Erasmus University Medical Center, Rotterdam
| |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to provide an update regarding the gut barrier and its involvement with chronic diseases, as well as to review biomarkers for identification of gut barrier integrity. This review is timely and relevant as our knowledge is increasing regarding the role of the gut microbiome and the gut barrier in health and disease. RECENT FINDINGS This review provides an overview of: the gut barrier, which is complex and comprised of the mucus layer and the intestinal apical junctional protein complex; the gut microbiome in its relation to regulating the integrity of the gut barrier; select acute and chronic conditions that are known to be associated with gut dysbiosis and impaired gut integrity or 'leaky gut'; and current means for identifying loss in gut barrier integrity. SUMMARY Many chronic conditions are associated with gut dysbiosis and systemic inflammation. Identifying whether the gut barrier is compromised in these conditions could help to inform potential therapeutics as a means to correct losses in gut barrier integrity and mitigate associated medical conditions.
Collapse
Affiliation(s)
- Anthony Santilli
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Stavros Stefanopoulos
- Department of General Surgery, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH
| | - Gail A.M. Cresci
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Department of Pediatric Gastroenterology, Hepatology & Nutrition, Pediatric Institute, Cleveland Clinic Children’s Hospital, Cleveland, OH
| |
Collapse
|
24
|
Harris LA, Cash BD, Moftah K, Franklin H. An Open-label, Multicenter Study to Assess the Efficacy and Safety of a Novel Probiotic Blend in Patients With Functional Gastrointestinal Symptoms. J Clin Gastroenterol 2022; 56:444-451. [PMID: 34028393 PMCID: PMC8989638 DOI: 10.1097/mcg.0000000000001567] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 04/09/2021] [Indexed: 12/19/2022]
Abstract
GOAL A novel 5-strain (Bl-04, Bi-07, HN019, NCFM, and Lpc-37) probiotic blend was developed and its safety and efficacy were evaluated in patients with functional gastrointestinal (GI) symptoms. BACKGROUND These strains administered together have not previously been investigated. STUDY Patients aged 18 to 75 years with functional GI symptoms were eligible for inclusion in a single-arm, open-label, multicenter study (NCT04155801). An oral capsule containing the novel probiotic blend was administered once daily for 30 days. The primary efficacy endpoint was patient-reported improvement in overall GI well-being at day 30. Secondary efficacy endpoints included changes in GI symptoms assessed using the GI Health Symptom Questionnaire. Incidence of treatment-emergent adverse events was recorded at all visits. RESULTS Of 188 enrolled patients, 72.3% were female and mean (SD) age was 44.1 (13.4) years. At day 30, 85.1% of patients achieved the primary endpoint, a positive response signifying improvement in overall GI well-being. Improvements from baseline were reported at day 30 in diarrhea frequency (baseline frequency≥3 to 4 d/wk) and severity (baseline severity≥5/10) for 75.8% and 87.3% of patients, respectively. Over the same time period, constipation frequency (baseline frequency≥3 to 4 d/wk) and severity (baseline severity≥5/10) improved in 73.6% and 80.4% of patients, respectively. Most patients reported improvements at day 30 in frequency and severity of straining, urgency, abdominal pain/discomfort, bloating, and distention. Improvements reported at day 30 were generally observable at day 14. No safety signals were identified. CONCLUSION A novel 5-strain probiotic blend improved functional GI symptoms and was safe.
Collapse
Affiliation(s)
- Lucinda A. Harris
- Division of Gastroenterology and Hepatology, Mayo Clinic, Alix School of Medicine, Scottsdale, AZ
| | - Brooks D. Cash
- Division of Gastroenterology, Hepatology, and Nutrition, University of Texas Health Science Center, Houston, TX
| | | | - Howard Franklin
- Salix Pharmaceuticals, A Division of Bausch Health US LLC, Bridgewater, NJ
| |
Collapse
|
25
|
Gut–Skin Axis: Unravelling the Connection between the Gut Microbiome and Psoriasis. Biomedicines 2022; 10:biomedicines10051037. [PMID: 35625774 PMCID: PMC9138548 DOI: 10.3390/biomedicines10051037] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/23/2022] [Accepted: 04/27/2022] [Indexed: 12/11/2022] Open
Abstract
Evidence has shown that gut microbiome plays a role in modulating the development of diseases beyond the gastrointestinal tract, including skin disorders such as psoriasis. The gut–skin axis refers to the bidirectional relationship between the gut microbiome and skin health. This is regulated through several mechanisms such as inflammatory mediators and the immune system. Dysregulation of microbiota has been seen in numerous inflammatory skin conditions such as atopic dermatitis, rosacea, and psoriasis. Understanding how gut microbiome are involved in regulating skin health may lead to development of novel therapies for these skin disorders through microbiome modulation, in particularly psoriasis. In this review, we will compare the microbiota between psoriasis patients and healthy control, explain the concept of gut–skin axis and the effects of gut dysbiosis on skin physiology. We will also review the current evidence on modulating gut microbiome using probiotics in psoriasis.
Collapse
|
26
|
Mousa WK, Chehadeh F, Husband S. Recent Advances in Understanding the Structure and Function of the Human Microbiome. Front Microbiol 2022; 13:825338. [PMID: 35185849 PMCID: PMC8851206 DOI: 10.3389/fmicb.2022.825338] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/11/2022] [Indexed: 12/11/2022] Open
Abstract
Trillions of microbes live within our bodies in a deep symbiotic relationship. Microbial populations vary across body sites, driven by differences in the environment, immunological factors, and interactions between microbial species. Major advances in genome sequencing enable a better understanding of microbiome composition. However, most of the microbial taxa and species of the human microbiome are still unknown. Without revealing the identity of these microbes as a first step, we cannot appreciate their role in human health and diseases. A shift in the microbial balance, termed dysbiosis, is linked to a broad range of diseases from simple colitis and indigestion to cancer and dementia. The last decade has witnessed an explosion in microbiome research that led to a better understanding of the microbiome structure and function. This understanding leads to potential opportunities to develop next-generation microbiome-based drugs and diagnostic biomarkers. However, our understanding is limited given the highly personalized nature of the microbiome and its complex and multidirectional interactions with the host. In this review, we discuss: (1) our current knowledge of microbiome structure and factors that shape the microbial composition, (2) recent associations between microbiome dysbiosis and diseases, and (3) opportunities of new microbiome-based therapeutics. We analyze common themes, promises, gaps, and challenges of the microbiome research.
Collapse
Affiliation(s)
- Walaa K. Mousa
- College of Pharmacy, Al Ain University of Science and Technology, Al Ain, United Arab Emirates
- Department of Biology, Whitman College, Walla Walla, WA, United States
- College of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Fadia Chehadeh
- Department of Biology, Whitman College, Walla Walla, WA, United States
| | - Shannon Husband
- Department of Biology, Whitman College, Walla Walla, WA, United States
| |
Collapse
|
27
|
Wang Y, Dan K, Xue X, Yang X, Feng X, Yang Q, Yang J, Chen B. Translocating lipopolysaccharide correlates with the severity of enterovirus A71-induced HFMD by promoting pro-inflammation and viral IRES activity. Gut Pathog 2021; 13:69. [PMID: 34809671 PMCID: PMC8607650 DOI: 10.1186/s13099-021-00465-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 11/09/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND The increase of inflammation-inducing enterobacteria was recently observed in severe hand, foot, and mouth disease (HFMD) caused by Enterovirus A71 (EV-A71). This study aimed to verify the occurrence of bacterial translocation (BT) and further explore the contributory role of BT to severity of EV-A71-mediated HFMD cases. METHODS Serum specimens from 65 mild and 65 severe EV-A71-associated HFMD cases and 65 healthy children were collected. EV-A71 VP1 in serum, inflammatory mediators including C-reactive protein, IL-1β, IL-6, interferon-γ and tumor necrosis factor-α, BT related biomarkers including Claudin-3, intestinal fatty acid binding protein, lipopolysaccharide (LPS), soluble CD14 (sCD14) and endotoxin core antibody were measured by ELISA. Bacterial DNA (BactDNA) fragments were quantified by quantified PCR (qPCR). Rhabdomyosarcoma (RD) or SH-SY5Y cells, infected with LPS-pre-incubated EV-A71 or transfected with plasmid containing viral 2Apro or mRNA containing viral internal ribosomal entry site (IRES), were post-treated with or without LPS in vitro. EV-A71 RNA and viral or cellular proteins were determined by qPCR and western blot, respectively. RESULTS Compared to mild HFMD patients, remarkably higher inflammatory mediators as well as BT-related biomarkers except BactDNA were observed in severe HFMD cases (all P < 0.05). In severe HFMD group, circulating concentrations of LPS and sCD14 showed statistical correlations with inflammation indices (all P < 0.05), serum levels of EV-A71 VP1 were found to be positively correlated with serum LPS (r = 0.341, P = 0.005) and serum sCD14 (r = 0.458, P < 0.001). In vitro, EV-A71 attachment and internalization were only slightly promoted by LPS pre-incubation; however, EV-A71 proliferation and viral 2Apro-mediated IRES activity were significantly accelerated by LPS post-treatment. CONCLUSIONS Our results collectively indicate that gut-derived translocating LPS contributes to the severity of EV-A71-induced HFMD by driving inflammatory response and viral proliferation via viral 2Apro-mediated IRES.
Collapse
Affiliation(s)
- Yuya Wang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Kena Dan
- Department of Dermatology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Xiaoling Xue
- Department of Hematology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Xiongbo Yang
- Department of Dermatology, Chongqing University Three Gorges Hospital, Chongqing, 404100, China
| | - Xujiao Feng
- Department of Infectious Diseases, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, 046000, China
| | - Qingqing Yang
- Department of Infectious Diseases, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, 046000, China
| | - Jing Yang
- Department of Dermatology, Chongqing University Three Gorges Hospital, Chongqing, 404100, China.
| | - Bangtao Chen
- Department of Dermatology, Chongqing University Three Gorges Hospital, Chongqing, 404100, China.
| |
Collapse
|
28
|
Hao Y, Zhu YJ, Zou S, Zhou P, Hu YW, Zhao QX, Gu LN, Zhang HZ, Wang Z, Li J. Metabolic Syndrome and Psoriasis: Mechanisms and Future Directions. Front Immunol 2021; 12:711060. [PMID: 34367173 PMCID: PMC8343100 DOI: 10.3389/fimmu.2021.711060] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/07/2021] [Indexed: 02/05/2023] Open
Abstract
Psoriasis is an immune-mediated systemic disease with associated comorbidities, including metabolic syndrome (MetS) which contributes substantially to premature mortality in patients with psoriasis. However, the pathological mechanisms underlying this comorbidity are unclear. Studies have shown that the pathological parameters of psoriasis mediate the development of MetS. We reviewed the potential mechanisms which mediate the association between psoriasis and MetS, including endoplasmic reticulum stress, pro-inflammatory cytokine releases, excess production of reactive oxygen species, alterations in adipocytokine levels and gut microbiota dysbiosis. Here, we highlight important research questions regarding this association and offer insights into MetS research and treatment.
Collapse
Affiliation(s)
- Yan Hao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Ya-Juan Zhu
- Department of Biotherapy and Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Song Zou
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| | - Pei Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Ya-Wen Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Qi-Xiang Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Lin-Na Gu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Hao-Zhou Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Zhen Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China.,Department of Liver Surgery & Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China.,Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Jiong Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| |
Collapse
|
29
|
Molecular Pathogenesis of Psoriasis and Biomarkers Reflecting Disease Activity. J Clin Med 2021; 10:jcm10153199. [PMID: 34361983 PMCID: PMC8346978 DOI: 10.3390/jcm10153199] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 07/18/2021] [Accepted: 07/19/2021] [Indexed: 12/17/2022] Open
Abstract
Psoriasis is a chronic inflammatory skin disease induced by multifactorial causes and is characterized by bothersome, scaly reddish plaques, especially on frequently chafed body parts, such as extensor sites of the extremities. The latest advances in molecular-targeted therapies using biologics or small-molecule inhibitors help to sufficiently treat even the most severe psoriatic symptoms and the extra cutaneous comorbidities of psoriatic arthritis. The excellent clinical effects of these therapies provide a deeper understanding of the impaired quality of life caused by this disease and the detailed molecular mechanism in which the interleukin (IL)-23/IL-17 axis plays an essential role. To establish standardized therapeutic strategies, biomarkers that define deep remission are indispensable. Several molecules, such as cytokines, chemokines, antimicrobial peptides, and proteinase inhibitors, have been recognized as potent biomarker candidates. In particular, blood protein markers that are repeatedly measurable can be extremely useful in daily clinical practice. Herein, we summarize the molecular mechanism of psoriasis, and we describe the functions and induction mechanisms of these biomarker candidates.
Collapse
|
30
|
Kozłowska D, Myśliwiec H, Harasim-Symbor E, Milewska AJ, Chabowski A, Flisiak I. Serum fatty acid binding protein 5 (FABP5) as a potential biomarker of inflammation in psoriasis. Mol Biol Rep 2021; 48:4421-4429. [PMID: 34131888 PMCID: PMC8260421 DOI: 10.1007/s11033-021-06461-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/29/2021] [Indexed: 12/16/2022]
Abstract
Fatty acid binding protein 5 (FABP5) is elevated in psoriatic keratinocytes and could be involved in systemic metabolic disturbances in psoriasis. The aim of the study was to evaluate serum FABP5 in obese and non-obese psoriatic patients, to assess the relationship between FABP5 and the duration, severity of the disease, inflammatory and metabolic markers and influence of treatment with narrowband-ultraviolet B (NB-UVB). Seventy-four patients (30 treated with NB-UVB) with psoriasis were enrolled in the study. The serum concentrations of FABP5 were measured using Human FABP5 Enzyme-Linked Immunosorbent Assay kit. Serum fatty acids were measured by gas-liquid chromatography. Serum FABP5 levels in psoriatic patients were higher versus control group (P < 0.001). FABP5 in patients with PASI > 20 was higher compared to the mild group (PASI < 10) (P < 0.001) and serum FABP5 correlated positively with PASI score (r = 0.41, P < 0.001). There was also positive correlation between FABP5 and basic inflammation indices. Decrease of PASI after NB-UVB treatment (P < 0.001) was observed and accompanied by decrease of the serum FABP5 (P = 0.007). FABP5 is a potential marker of psoriasis, its severity and clinical outcome after therapy with NB-UVB. FABP5 may reflect metabolic disturbances in psoriatic patients.
Collapse
Affiliation(s)
- Dorota Kozłowska
- Department of Dermatology and Venereology, Medical University of Bialystok, Żurawia str. 14, 15-540, Białystok, Poland.
| | - Hanna Myśliwiec
- Department of Dermatology and Venereology, Medical University of Bialystok, Żurawia str. 14, 15-540, Białystok, Poland
| | - Ewa Harasim-Symbor
- Department of Physiology, Medical University of Bialystok, Białystok, Poland
| | - Anna Justyna Milewska
- Department of Statistics and Medical Informatics, Medical University of Bialystok, Bialystok, Poland
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, Białystok, Poland
| | - Iwona Flisiak
- Department of Dermatology and Venereology, Medical University of Bialystok, Żurawia str. 14, 15-540, Białystok, Poland
| |
Collapse
|
31
|
Trimethylamine N-Oxide, a Gut Microbiota-Derived Metabolite, Is Associated with Cardiovascular Risk in Psoriasis: A Cross-Sectional Pilot Study. Dermatol Ther (Heidelb) 2021; 11:1277-1289. [PMID: 33983475 PMCID: PMC8322249 DOI: 10.1007/s13555-021-00547-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 04/30/2021] [Indexed: 02/07/2023] Open
Abstract
Introduction Trimethylamine N-oxide (TMAO), a gut microbiota metabolite from dietary phosphatidylcholine, is involved in the pathogenesis of atherosclerosis and cardiovascular diseases. Psoriasis is associated with increased cardiovascular risk that is not captured by traditional biomarkers. The aim of the present study was to assess TMAO concentration in psoriasis and evaluate the relationship between TMAO and cardiovascular risk in psoriatic patients. Methods In 72 patients with psoriasis and 40 age- and sex-matched non-psoriatic controls, we evaluated fasting plasma TMAO, measured by high-performance liquid chromatography, and cardiovascular risk assessed by various scoring systems such as Framingham, QRISK2, AHA/ACC, and Reynolds risk scores. Results In patients with psoriasis, TMAO concentration was significantly higher than in the control group (195.68 [133.54–332.58] ng/ml versus 126.06 [84.29–156.88] ng/ml, respectively; p < 0.001). Plasma TMAO concentration was significantly correlated with age, total cholesterol, triglycerides, systolic and diastolic blood pressure. Furthermore, the receiver-operating characteristic (ROC) and multiple regression analysis showed that TMAO is an independent predictor of cardiovascular risk. Conclusion TMAO is a valuable candidate for biomarker and a translational link between dysbiosis and atherosclerosis in psoriasis.
Collapse
|
32
|
Psoriasis and Gut Microbiome-Current State of Art. Int J Mol Sci 2021; 22:ijms22094529. [PMID: 33926088 PMCID: PMC8123672 DOI: 10.3390/ijms22094529] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 12/12/2022] Open
Abstract
Psoriasis is a chronic, immune-mediated inflammatory disease that affects around 125 million people worldwide. Several studies concerning the gut microbiota composition and its role in disease pathogenesis recently demonstrated significant alterations among psoriatic patients. Certain parameters such as Firmicutes/Bacteroidetes ratio or Psoriasis Microbiome Index were developed in order to distinguish between psoriatic and healthy individuals. The “leaky gut syndrome” and bacterial translocation is considered by some authors as a triggering factor for the onset of the disease, as it promotes chronic systemic inflammation. The alterations were also found to resemble those in inflammatory bowel diseases, obesity and certain cardiovascular diseases. Microbiota dysbiosis, depletion in SCFAs production, increased amount of produced TMAO, dysregulation of the pathways affecting the balance between lymphocytes populations seem to be the most significant findings concerning gut physiology in psoriatic patients. The gut microbiota may serve as a potential response-to-treatment biomarker in certain cases of biological treatment. Oral probiotics administration as well as fecal microbial transplantation were most reported in bringing health benefits to psoriatic patients. However, the issue of psoriatic bacterial gut composition, its role and healing potential needs further investigation. Here we reviewed the literature on the current state of the relationship between psoriasis and gut microbiome.
Collapse
|
33
|
Olejniczak-Staruch I, Ciążyńska M, Sobolewska-Sztychny D, Narbutt J, Skibińska M, Lesiak A. Alterations of the Skin and Gut Microbiome in Psoriasis and Psoriatic Arthritis. Int J Mol Sci 2021; 22:ijms22083998. [PMID: 33924414 PMCID: PMC8069836 DOI: 10.3390/ijms22083998] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/05/2021] [Accepted: 04/06/2021] [Indexed: 12/12/2022] Open
Abstract
Numerous scientific studies in recent years have shown significant skin and gut dysbiosis among patients with psoriasis. A significant decrease in microbiome alpha-diversity (abundance of different bacterial taxa measured in one sample) as well as beta-diversity (microbial diversity in different samples) was noted in psoriasis skin. It has been proven that the representation of Cutibacterium, Burkholderia spp., and Lactobacilli is decreased and Corynebacterium kroppenstedii, Corynebacterium simulans, Neisseria spp., and Finegoldia spp. increased in the psoriasis skin in comparison to healthy skin. Alterations in the gut microbiome in psoriasis are similar to those observed in patients with inflammatory bowel disease. In those two diseases, the F. prausnitzii, Bifidobacterium spp., Lactobacillus spp., Parabacteroides and Coprobacillus were underrepresented, while the abundance of Salmonella sp., Campylobacter sp., Helicobacter sp., Escherichia coli, Alcaligenes sp., and Mycobacterium sp. was increased. Several research studies provided evidence for the significant influence of psoriasis treatments on the skin and gut microbiome and a positive influence of orally administered probiotics on the course of this dermatosis. Further research is needed to determine the influence of the microbiome on the development of inflammatory skin diseases. The changes in microbiome under psoriasis treatment can serve as a potential biomarker of positive response to the administered therapy.
Collapse
Affiliation(s)
- Irmina Olejniczak-Staruch
- Department of Dermatology, Pediatric Dermatology and Dermatological Oncology, Medical University of Lodz, 91-347 Lodz, Poland; (D.S.-S.); (J.N.); (M.S.); (A.L.)
- Dermoklinika Centrum Medyczne, 90-436 Lodz, Poland
- Correspondence: ; Tel.: +48-42-230-9657
| | - Magdalena Ciążyńska
- Department of Proliferative Diseases, Nicolaus Copernicus Multidisciplinary Centre for Oncology and Traumatology, 93-513 Lodz, Poland;
| | - Dorota Sobolewska-Sztychny
- Department of Dermatology, Pediatric Dermatology and Dermatological Oncology, Medical University of Lodz, 91-347 Lodz, Poland; (D.S.-S.); (J.N.); (M.S.); (A.L.)
| | - Joanna Narbutt
- Department of Dermatology, Pediatric Dermatology and Dermatological Oncology, Medical University of Lodz, 91-347 Lodz, Poland; (D.S.-S.); (J.N.); (M.S.); (A.L.)
| | - Małgorzata Skibińska
- Department of Dermatology, Pediatric Dermatology and Dermatological Oncology, Medical University of Lodz, 91-347 Lodz, Poland; (D.S.-S.); (J.N.); (M.S.); (A.L.)
| | - Aleksandra Lesiak
- Department of Dermatology, Pediatric Dermatology and Dermatological Oncology, Medical University of Lodz, 91-347 Lodz, Poland; (D.S.-S.); (J.N.); (M.S.); (A.L.)
| |
Collapse
|
34
|
Colucci R, Moretti S. Implication of Human Bacterial Gut Microbiota on Immune-Mediated and Autoimmune Dermatological Diseases and Their Comorbidities: A Narrative Review. Dermatol Ther (Heidelb) 2021; 11:363-384. [PMID: 33507493 PMCID: PMC8018919 DOI: 10.1007/s13555-021-00485-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Indexed: 12/12/2022] Open
Abstract
During the last decade, the advent of modern sequencing methods (next generation techniques, NGS) has helped describe the composition of the human gut microbiome, enabling us to understand the main characteristics of a healthy gut microbiome and, conversely, the magnitude of its disease-related changes. This new knowledge has revealed that healthy gut microbiota allow the maintenance of several crucial physiological functions, such as the ability to regulate the innate and adaptive immune systems. Increasing evidence has pointed out a condition of dysbiosis in several autoimmune/immune mediated dermatological conditions and specific gut microbial signatures have also been reported to correlate with clinical and prognostic parameters of such diseases. Based on a literature search of relevant published articles, this review debates the current knowledge and the possible pathogenic implications of bacterial gut microbiota composition assessed through NGS techniques in systemic lupus erythematosus, atopic dermatitis, psoriasis, and alopecia areata. Evidence of a potential role of specific gut microbiota signatures in modulating the clinical course of such diseases and their main comorbidities has been also reviewed.
Collapse
Affiliation(s)
- Roberta Colucci
- Section of Dermatology, Department of Health Sciences, University of Florence, Florence, Italy.
| | - Silvia Moretti
- Section of Dermatology, Department of Health Sciences, University of Florence, Florence, Italy
| |
Collapse
|
35
|
Wu P, Ye H, Cai X, Li C, Li S, Chen M, Wang M, Heidari AA, Chen M, Li J, Chen H, Huang X, Wang L. An Effective Machine Learning Approach for Identifying Non-Severe and Severe Coronavirus Disease 2019 Patients in a Rural Chinese Population: The Wenzhou Retrospective Study. IEEE ACCESS : PRACTICAL INNOVATIONS, OPEN SOLUTIONS 2021; 9:45486-45503. [PMID: 34786313 PMCID: PMC8545214 DOI: 10.1109/access.2021.3067311] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 03/15/2021] [Indexed: 06/13/2023]
Abstract
This paper has proposed an effective intelligent prediction model that can well discriminate and specify the severity of Coronavirus Disease 2019 (COVID-19) infection in clinical diagnosis and provide a criterion for clinicians to weigh scientific and rational medical decision-making. With indicators as the age and gender of the patients and 26 blood routine indexes, a severity prediction framework for COVID-19 is proposed based on machine learning techniques. The framework consists mainly of a random forest and a support vector machine (SVM) model optimized by a slime mould algorithm (SMA). When the random forest was used to identify the key factors, SMA was employed to train an optimal SVM model. Based on the COVID-19 data, comparative experiments were conducted between RF-SMA-SVM and several well-known machine learning algorithms performed. The results indicate that the proposed RF-SMA-SVM not only achieves better classification performance and higher stability on four metrics, but also screens out the main factors that distinguish severe COVID-19 patients from non-severe ones. Therefore, there is a conclusion that the RF-SMA-SVM model can provide an effective auxiliary diagnosis scheme for the clinical diagnosis of COVID-19 infection.
Collapse
Affiliation(s)
- Peiliang Wu
- Department of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhou325000China
| | - Hua Ye
- Department of Pulmonary and Critical Care MedicineAffiliated Yueqing Hospital, Wenzhou Medical UniversityYueqing325600China
| | - Xueding Cai
- Department of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhou325000China
| | - Chengye Li
- Department of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhou325000China
| | - Shimin Li
- College of Computer Science and Artificial IntelligenceWenzhou UniversityWenzhou325035China
| | - Mengxiang Chen
- Department of Information TechnologyWenzhou Vocational College of Science and TechnologyWenzhou325006China
| | - Mingjing Wang
- College of Computer Science and Artificial IntelligenceWenzhou UniversityWenzhou325035China
| | - Ali Asghar Heidari
- School of Surveying and Geospatial Engineering, College of EngineeringUniversity of TehranTehran1417466191Iran
- Department of Computer ScienceSchool of ComputingNational University of SingaporeSingapore117417
| | - Mayun Chen
- Department of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhou325000China
| | - Jifa Li
- Department of Pulmonary and Critical Care MedicineAffiliated Yueqing Hospital, Wenzhou Medical UniversityYueqing325600China
| | - Huiling Chen
- College of Computer Science and Artificial IntelligenceWenzhou UniversityWenzhou325035China
| | - Xiaoying Huang
- Department of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhou325000China
| | - Liangxing Wang
- Department of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhou325000China
| |
Collapse
|
36
|
Vermandere K, Bostick RM, Tran HQ, Gewirtz AT, Barry EL, Rutherford RE, Seabrook ME, Fedirko V. Effects of Supplemental Calcium and Vitamin D on Circulating Biomarkers of Gut Barrier Function in Patients with Colon Adenoma: A Randomized Clinical Trial. Cancer Prev Res (Phila) 2021; 14:393-402. [PMID: 33229339 PMCID: PMC8137511 DOI: 10.1158/1940-6207.capr-20-0461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/22/2020] [Accepted: 11/11/2020] [Indexed: 11/16/2022]
Abstract
Gut barrier dysfunction promotes chronic inflammation, contributing to several gastrointestinal diseases, including colorectal cancer. Preliminary evidence suggests that vitamin D and calcium could prevent colorectal carcinogenesis, in part, by influencing gut barrier function. However, relevant human data are scarce. We tested the effects of supplemental calcium (1,200 mg/day) and/or vitamin D3 (1,000 IU/day) on circulating concentrations of biomarkers of gut permeability (anti-flagellin and anti-lipopolysaccharide IgA and IgG, measured via ELISA) from baseline to 1 and 3 or 5 years postbaseline among 175 patients with colorectal adenoma in a randomized, double-blinded, placebo-controlled clinical trial. We also assessed factors associated with baseline concentrations of these biomarkers. We found no appreciable effects of supplemental vitamin D3 and/or calcium on individual or aggregate biomarkers of gut permeability. At baseline, a combined permeability score (the summed concentrations of all four biomarkers) was 14% lower among women (P = 0.01) and 10% higher among those who consumed >1 serving per day of red or processed meats relative to those who consumed none (P trend = 0.03). The permeability score was estimated to be 49% higher among participants with a body mass index (BMI) > 35 kg/m2 relative to those with a BMI < 22.5 kg/m2 (P trend = 0.17). Our results suggest that daily supplemental vitamin D3 and/or calcium may not modify circulating concentrations of gut permeability biomarkers within 1 or 3-5 years, but support continued investigation of modifiable factors, such as diet and excess adiposity, that could affect gut permeability. PREVENTION RELEVANCE: Calcium and vitamin D may be involved in regulating and maintaining the integrity of the intestinal mucosal barrier, the dysfunction of which results in exposure of the host to luminal bacteria, endotoxins, and antigens leading to potentially cancer-promoting endotoxemia and chronic colon inflammation. While our results suggest that daily supplementation with these chemopreventive agents does not modify circulating concentrations of gut permeability biomarkers, they support continued investigation of other potential modifiable factors, such as diet and excess adiposity, that could alter gut barrier function, to inform the development of treatable biomarkers of risk for colorectal neoplasms and effective colon cancer preventive strategies.
Collapse
Affiliation(s)
- Kelly Vermandere
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Roberd M Bostick
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
- Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Hao Q Tran
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia
| | - Andrew T Gewirtz
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia
| | - Elizabeth L Barry
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Robin E Rutherford
- Division of Digestive Diseases, Department of Medicine, Emory University, Atlanta, Georgia
| | | | - Veronika Fedirko
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia.
- Winship Cancer Institute, Emory University, Atlanta, Georgia
| |
Collapse
|
37
|
De Pessemier B, Grine L, Debaere M, Maes A, Paetzold B, Callewaert C. Gut-Skin Axis: Current Knowledge of the Interrelationship between Microbial Dysbiosis and Skin Conditions. Microorganisms 2021; 9:353. [PMID: 33670115 PMCID: PMC7916842 DOI: 10.3390/microorganisms9020353] [Citation(s) in RCA: 252] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/25/2021] [Accepted: 02/07/2021] [Indexed: 02/06/2023] Open
Abstract
The microbiome plays an important role in a wide variety of skin disorders. Not only is the skin microbiome altered, but also surprisingly many skin diseases are accompanied by an altered gut microbiome. The microbiome is a key regulator for the immune system, as it aims to maintain homeostasis by communicating with tissues and organs in a bidirectional manner. Hence, dysbiosis in the skin and/or gut microbiome is associated with an altered immune response, promoting the development of skin diseases, such as atopic dermatitis, psoriasis, acne vulgaris, dandruff, and even skin cancer. Here, we focus on the associations between the microbiome, diet, metabolites, and immune responses in skin pathologies. This review describes an exhaustive list of common skin conditions with associated dysbiosis in the skin microbiome as well as the current body of evidence on gut microbiome dysbiosis, dietary links, and their interplay with skin conditions. An enhanced understanding of the local skin and gut microbiome including the underlying mechanisms is necessary to shed light on the microbial involvement in human skin diseases and to develop new therapeutic approaches.
Collapse
Affiliation(s)
- Britta De Pessemier
- Center for Microbial Ecology and Technology, Ghent University, Coupure Links 653, 9000 Ghent, Belgium; (B.D.P.); (M.D.); (A.M.)
| | - Lynda Grine
- Department of Head & Skin, Ghent University, Corneel Heymanslaan 10, 9000 Ghent, Belgium;
| | - Melanie Debaere
- Center for Microbial Ecology and Technology, Ghent University, Coupure Links 653, 9000 Ghent, Belgium; (B.D.P.); (M.D.); (A.M.)
| | - Aglaya Maes
- Center for Microbial Ecology and Technology, Ghent University, Coupure Links 653, 9000 Ghent, Belgium; (B.D.P.); (M.D.); (A.M.)
| | | | - Chris Callewaert
- Center for Microbial Ecology and Technology, Ghent University, Coupure Links 653, 9000 Ghent, Belgium; (B.D.P.); (M.D.); (A.M.)
| |
Collapse
|
38
|
Dysregulation of the gut-brain-skin axis and key overlapping inflammatory and immune mechanisms of psoriasis and depression. Biomed Pharmacother 2021; 137:111065. [PMID: 33540138 DOI: 10.1016/j.biopha.2020.111065] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/14/2020] [Accepted: 11/20/2020] [Indexed: 12/21/2022] Open
Abstract
The occurrence, progression and recurrence of psoriasis are thought to be related to mood and psychological disorders such as depression. Psoriasis can lead to depression, and depression, in turn, exacerbates psoriasis. No specific mechanism can explain the association between psoriasis and depression. The gut-brain-skin axis has been used to explain correlations among the gut microbiota, emotional states and systemic and skin inflammation, and this axis may be associated with overlapping mechanisms between psoriasis and depression. Therefore, in the context of the gut-brain-skin axis, we systematically summarized and comparatively analysed the inflammatory and immune mechanisms of psoriasis and depression and illustrated the dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis and the gut microbiota. This review provides a theoretical basis and new targets for the treatment of psoriasis and depression.
Collapse
|
39
|
Sikora M, Stec A, Chrabaszcz M, Giebultowicz J, Samborowska E, Jazwiec R, Dadlez M, Olszewska M, Rudnicka L. Clinical Implications of Intestinal Barrier Damage in Psoriasis. J Inflamm Res 2021; 14:237-243. [PMID: 33542642 PMCID: PMC7851376 DOI: 10.2147/jir.s292544] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 12/24/2020] [Indexed: 12/12/2022] Open
Abstract
Background An increasing amount of evidence suggests an association between increased intestinal permeability and the pathogenesis of chronic inflammatory diseases. However, the clinical significance of gut barrier dysfunction in psoriasis remains to be established. Objective To evaluate whether there are differences in disease activity, the severity of gastrointestinal symptoms and the blood concentration of bacterial metabolites in psoriatic patients with a normal and altered intestinal barrier. Patients and Methods Gut barrier integrity was assessed with the serum concentrations of claudin-3, a modulator of intestinal tight junctions and an intestinal fatty acid-binding protein, a marker of enterocyte damage. Gastrointestinal symptoms were evaluated with a validated questionnaire. The concentration of trimethylamine N-oxide (TMAO), a gut microbiota-associated metabolite, was measured with high-performance liquid chromatography. Results One hundred and fourteen patients with psoriasis were finally enrolled in the study – 68 with an altered gut barrier and 46 with a properly functioning intestinal barrier. Patients with an altered gut barrier showed a significantly higher score in the Gastrointestinal Symptom Rating Scale (3.20 vs 1.46, p<0.001). Moreover, patients with psoriasis and a disrupted intestinal barrier demonstrated a higher disease activity (PASI: 19.7 vs 10.3, p<0.001) and systemic inflammatory parameters (neutrophil-to-lymphocyte ratio: 2.86 vs 1.71, p<0.001; C-reactive protein 3.76 vs 1.92; p<0.05). The marker of bacterial translocation was significantly higher in psoriatic patients with damaged gut integrity (TMAO: 375.7±51.9 vs 119.4±27.5 ng/mL; p<0.05). Conclusion The altered gut barrier in psoriasis is associated with gastrointestinal symptoms, systemic inflammatory profile and the increased blood concentration of gut microbiota-derived metabolite – TMAO. Intestinal barrier modulation represents a new promising therapeutic approach.
Collapse
Affiliation(s)
- Mariusz Sikora
- Department of Dermatology, Medical University of Warsaw, Warsaw, Poland
| | - Albert Stec
- Department of Dermatology, Medical University of Warsaw, Warsaw, Poland
| | | | - Joanna Giebultowicz
- Department of Bioanalysis and Drug Analysis, Faculty of Pharmacy with the Laboratory Medicine Division, Medical University of Warsaw, Warsaw, Poland
| | - Emilia Samborowska
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Radoslaw Jazwiec
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Michal Dadlez
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland.,Institute of Genetics and Biotechnology, Biology Department, Warsaw University, Warsaw, Poland
| | | | - Lidia Rudnicka
- Department of Dermatology, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
40
|
El-Hakim Y, Mani KK, Eldouh A, Pandey S, Grimaldo MT, Dabney A, Pilla R, Sohrabji F. Sex differences in stroke outcome correspond to rapid and severe changes in gut permeability in adult Sprague-Dawley rats. Biol Sex Differ 2021; 12:14. [PMID: 33451354 PMCID: PMC7811247 DOI: 10.1186/s13293-020-00352-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 12/21/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Sex differences in experimental stroke outcomes are well documented, such that adult males have a greater infarct volume, increased stroke-induced mortality, and more severe sensory-motor impairment. Based on recent evidence that the gut is an early responder to stroke, the present study tested the hypothesis that sex differences in stroke severity will be accompanied by rapid and greater permeability of the gut-blood barrier and gut dysbiosis in males as compared to females. METHOD Male and female Sprague-Dawley rats (5-7 months of age) were subject to endothelin (ET)-1-induced middle cerebral artery occlusion (MCAo). Sensory-motor tests were conducted pre- and 2 days after MCAo. Gut permeability was assessed in serum samples using biomarkers of gut permeability as well as functional assays using size-graded dextrans. Histological analysis of the gut was performed with H&E staining, periodic acid-Schiff for mucus, and immunohistochemistry for the tight junction protein, ZO-1. Fecal samples obtained pre- and post-stroke were analyzed for bacterial taxa and short-chain fatty acids (SCFAs). RESULTS After stroke, males displayed greater mortality, worse sensory-motor deficit, and higher serum levels of proinflammatory cytokines IL-17A, MCP-1, and IL-5 as compared to females. MCAo-induced gut permeability was rapid and severe in males as indicated by dextran extravasation from the gut to the blood in the hyperacute (< 2 h) and early acute (2 days) phase of stroke. This was accompanied by dysmorphology of the gut villi and dysregulation of the tight junction protein ZO-1 in the acute phase. Fecal 16s sequencing showed no differences in bacterial diversity in the acute phase of stroke. Predictive modeling indicated that markers of gut permeability were associated with acute sensory-motor impairment and infarct volume. CONCLUSIONS These data show that extensive leakiness of the gut barrier is associated with severe post-stroke disability and suggest that reinforcing this barrier may improve stroke outcomes.
Collapse
Affiliation(s)
- Yumna El-Hakim
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University-Health, 8447 Riverside Pkwy, Bryan, TX, 77807, USA
| | - Kathiresh Kumar Mani
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University-Health, 8447 Riverside Pkwy, Bryan, TX, 77807, USA
| | - Amir Eldouh
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University-Health, 8447 Riverside Pkwy, Bryan, TX, 77807, USA
| | - Sivani Pandey
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University-Health, 8447 Riverside Pkwy, Bryan, TX, 77807, USA
| | - Maria T Grimaldo
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University-Health, 8447 Riverside Pkwy, Bryan, TX, 77807, USA
| | - Alan Dabney
- Department of Statistics, College of Science, College Station, USA
| | - Rachel Pilla
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Texas A&M University, College Station, TX, USA
| | - Farida Sohrabji
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University-Health, 8447 Riverside Pkwy, Bryan, TX, 77807, USA.
| |
Collapse
|
41
|
Wang Y, Ding L, Yang J, Liu L, Dong L. Intestinal fatty acid-binding protein, a biomarker of intestinal barrier dysfunction, increases with the progression of type 2 diabetes. PeerJ 2021; 9:e10800. [PMID: 33604184 PMCID: PMC7863777 DOI: 10.7717/peerj.10800] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 12/29/2020] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE To investigate serum intestinal fatty acid-binding protein (I-FABP) in two groups of patients with different duration of hyperglycemia in a cross-sectional study. MATERIALS AND METHODS In the present study, a total of 280 individuals (158 outpatients and 122 inpatients) suffering from hyperglycemia were recruited between May and September 2019. The clinical information of all participants was collected from the hospital information system, including the duration of hyperglycemia, age, gender, hemoglobin A1c (HbA1c), 75-g oral glucose tolerance test including fasting plasma glucose (FPG), 2-hour plasma glucose (2hPG), fasting C-peptide (FC-pep), 2-hour C-peptide (2hC-pep), fasting insulin (FIns), and 2-hour insulin (2hIns). In addition, the morbidity of diabetic complications (retinopathy, neuropathy, and nephropathy) in the inpatient group was determined. Furthermore, the difference between 2hPG and FPG (ΔPG), the difference between 2hC-pep and FC-pep (ΔC-pep), and the difference between 2hIns and FIns (ΔIns) were calculated. The level of serum I-FABP, a biomarker of intestinal barrier (IB) dysfunction, was estimated by an enzyme-linked immunosorbent assay. RESULTS For the outpatient group, the median duration of hyperglycemia was less than a year; the serum I-FABP level was positively correlated with age (R = 0.299, P < 0.001). For the inpatient group, the median duration of hyperglycemia was ten years; correlation analysis showed that the serum I-FABP level was positively associated with age and ΔPG (R = 0.286, P = 0.001; R = 0.250, P = 0.006, respectively) while negatively associated with FC-pep and 2hC-pep (R = - 0.304, P = 0.001; R = - 0.241, P = 0.008, respectively); multiple linear regression analysis showed that the serum I-FABP level was positively associated with the duration of hyperglycemia (β = 0.362, P < 0.001); moreover, patients with retinopathy had a significantly higher I-FABP level than those without retinopathy (P = 0.001). CONCLUSIONS In the outpatients whose duration of hyperglycemia was less than a year, the serum I-FABP level was positively associated with age. In the inpatients with different courses of diabetes, the serum I-FABP level was positively associated with the duration of hyperglycemia and glycemic variability but negatively associated with islet beta-cell function; moreover, the serum I-FABP level was higher in patients with retinopathy than in those without retinopathy, suggesting that the IB dysfunction got worse with the progression of diabetes.
Collapse
Affiliation(s)
- Yifeng Wang
- Department of Emergency and Critical Care Medicine, Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Critical Care Medicine, Wuxi People’s Hospital affiliated to Nanjing Medical University, Wuxi, China
| | - Licheng Ding
- Department of Emergency Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Jiayue Yang
- Department of Endocrinology and Metabolism, Wuxi People’s Hospital affiliated to Nanjing Medical University, Wuxi, China
| | - Lijun Liu
- Department of Emergency and Critical Care Medicine, Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Liang Dong
- Department of Critical Care Medicine, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| |
Collapse
|
42
|
Chen L, Li J, Zhu W, Kuang Y, Liu T, Zhang W, Chen X, Peng C. Skin and Gut Microbiome in Psoriasis: Gaining Insight Into the Pathophysiology of It and Finding Novel Therapeutic Strategies. Front Microbiol 2020; 11:589726. [PMID: 33384669 PMCID: PMC7769758 DOI: 10.3389/fmicb.2020.589726] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022] Open
Abstract
Psoriasis affects the health of myriad populations around the world. The pathogenesis is multifactorial, and the exact driving factor remains unclear. This condition arises from the interaction between hyperproliferative keratinocytes and infiltrating immune cells, with poor prognosis and high recurrence. Better clinical treatments remain to be explored. There is much evidence that alterations in the skin and intestinal microbiome play an important role in the pathogenesis of psoriasis, and restoration of the microbiome is a promising preventive and therapeutic strategy for psoriasis. Herein, we have reviewed recent studies on the psoriasis-related microbiome in an attempt to confidently identify the “core” microbiome of psoriasis patients, understand the role of microbiome in the pathogenesis of psoriasis, and explore new therapeutic strategies for psoriasis through microbial intervention.
Collapse
Affiliation(s)
- Lihui Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
| | - Jie Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
| | - Wu Zhu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
| | - Yehong Kuang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
| | - Tao Liu
- Central Laboratory, Shenzhen Center for Chronic Disease Control and Prevention, Shenzhen, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
| | - Cong Peng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
| |
Collapse
|
43
|
Jaganjac M, Milkovic L, Gegotek A, Cindric M, Zarkovic K, Skrzydlewska E, Zarkovic N. The relevance of pathophysiological alterations in redox signaling of 4-hydroxynonenal for pharmacological therapies of major stress-associated diseases. Free Radic Biol Med 2020; 157:128-153. [PMID: 31756524 DOI: 10.1016/j.freeradbiomed.2019.11.023] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/04/2019] [Accepted: 11/17/2019] [Indexed: 02/07/2023]
Abstract
Modern analytical methods combined with the modern concepts of redox signaling revealed 4-hydroxy-2-nonenal (4-HNE) as particular growth regulating factor involved in redox signaling under physiological and pathophysiological circumstances. In this review current knowledge of the relevance of 4-HNE as "the second messenger of reactive oxygen species" (ROS) in redox signaling of representative major stress-associated diseases is briefly summarized. The findings presented allow for 4-HNE to be considered not only as second messenger of ROS, but also as one of fundamental factors of the stress- and age-associated diseases. While standard, even modern concepts of molecular medicine and respective therapies in majority of these diseases target mostly the disease-specific symptoms. 4-HNE, especially its protein adducts, might appear to be the bioactive markers that would allow better monitoring of specific pathophysiological processes reflecting their complexity. Eventually that could help development of advanced integrative medicine approach for patients and the diseases they suffer from on the personalized basis implementing biomedical remedies that would optimize beneficial effects of ROS and 4-HNE to prevent the onset and progression of the illness, perhaps even providing the real cure.
Collapse
Affiliation(s)
- Morana Jaganjac
- Qatar Analytics & BioResearch Lab, Anti Doping Lab Qatar, Sport City Street, Doha, Qatar
| | - Lidija Milkovic
- Rudjer Boskovic Institute, Laboratory for Oxidative Stress, Div. of Molecular Medicine, Bijenicka 54, Zagreb, Croatia
| | - Agnieszka Gegotek
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222, Bialystok, Poland
| | - Marina Cindric
- University of Zagreb, School of Medicine, Div. of Pathology, University Hospital Centre Zagreb, Kispaticeva 12, Zagreb, Croatia
| | - Kamelija Zarkovic
- University of Zagreb, School of Medicine, Div. of Pathology, University Hospital Centre Zagreb, Kispaticeva 12, Zagreb, Croatia
| | - Elzbieta Skrzydlewska
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222, Bialystok, Poland
| | - Neven Zarkovic
- Rudjer Boskovic Institute, Laboratory for Oxidative Stress, Div. of Molecular Medicine, Bijenicka 54, Zagreb, Croatia.
| |
Collapse
|
44
|
Barekatain R, Howarth GS, Willson NL, Cadogan D, Wilkinson S. Excreta biomarkers in response to different gut barrier dysfunction models and probiotic supplementation in broiler chickens. PLoS One 2020; 15:e0237505. [PMID: 32790727 PMCID: PMC7425878 DOI: 10.1371/journal.pone.0237505] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 07/28/2020] [Indexed: 12/13/2022] Open
Abstract
Increased intestinal permeability (IP) and inflammation are both linked with functionality of the intestinal barrier and in particular enterocytes. Currently, almost all assessment methods of the intestinal barrier function are invasive. The present study aimed to quantify selected proteins as novel biomarkers in excreta of broiler chickens to facilitate non-invasive assessment of gut barrier function using enzyme-linked immunosorbent assays (ELISA). It was further hypothesised that probiotics as feed additives may counteract gut barrier dysfunction. A 3 × 2 factorial arrangement of treatments was used with the main factors being gut barrier dysfunction models (control, rye-based diet, and dexamethasone-DEX) with and without probiotic supplementation (a three-strain Bacillus) using 72 male Ross 308 day-old chickens. Each of the 6 experimental treatments was replicated 12 times. On d 21 of age, fluorescein isothiocyanate dextran (FITC-d) uptake into serum was examined to test IP. Fresh excreta samples were collected on d 20. The biomarkers included alpha-1 antitrypsin (A1AT), intestinal fatty acid binding protein (I-FABP), lipocalin-2 (LCN2), fibronectin (FN), intestinal alkaline phosphatase (IAP), ovotransferrin (OVT) and superoxide dismutase [Cu-Zn] (SOD1). Only DEX increased (P<0.001) FITC-d passage to the blood on d 21 of age, indicating a greater IP. The excreta concentrations of A1AT, I-FABP and SOD1 were unaltered by the experimental treatments. DEX increased (P<0.05) FN concentration in excreta compared with control birds. Conversely, inclusion of rye in the diet reduced (P<0.05) FN but increased (P<0.001) OVT in excreta. Independently, DEX decreased IAP (P<0.05) in excreta compared with control and rye-fed birds. The excreta concentration of LCN2 tended (P = 0.086) to increase in birds injected by DEX. There was no demonstrable effect of probiotic addition on any of the studied parameters. Among the tested biomarkers, FN, IAP, and LCN2 revealed promise as biomarkers of intestinal barrier function quantified by ELISA kits.
Collapse
Affiliation(s)
- Reza Barekatain
- South Australian Research and Development Institute, Roseworthy, SA, Australia
- School of Animal and Veterinary Sciences, University of Adelaide, Roseworthy, SA, Australia
| | - Gordon S. Howarth
- School of Animal and Veterinary Sciences, University of Adelaide, Roseworthy, SA, Australia
| | - Nicky-Lee Willson
- School of Animal and Veterinary Sciences, University of Adelaide, Roseworthy, SA, Australia
| | | | | |
Collapse
|
45
|
Абатуров А, Никулина А. Antibiotic Therapy as a Risk Factor of Obesity Development in Children. ПЕДИАТРИЯ. ВОСТОЧНАЯ ЕВРОПА 2020:268-290. [DOI: 10.34883/pi.2020.8.2.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Настоящий обзор научной литературы посвящен вопросам, связанным с механизмами антибактериально-индуцированного адипогенеза. Антибиотиками, наиболее высоко ассоциированными с развитием ожирения у детей, считают: амоксициллин, цефотаксим, макролиды, тетрациклины, ванкомицин. На основании результатов филогенетических, метагеномных исследований эффектов антибиотиков установлено, что их применение в антенатальном, раннем постнатальном периоде приводит к пролонгированным изменениям как состава, так и функционирования микробиома, которые ассоциированы с повышенным риском последующего увеличения массы тела ребенка. Механизмы непосредственного влияния антибиотиков на адипогенез связаны с их способностью повышать аппетит за счет стимуляции высвобождения орексина и меланин-концентрирующего гормона; увеличивать абсорбцию пищевых ингредиентов; активировать липогенез; индуцировать митохондриальную дисфункцию и тем самым способствовать накоплению жирных кислот. Применение антибиотиков существенно изменяет структуру микробиома кишечника, а именно: развитие ожирения связано с высоким уровнем представительства бактерий филюмов Actinobacteria и Firmicutes в сочетании со снижением численности бактерий Bacteroidetes, Verrucomicrobia и Faecalibacterium prausnitzii. Антибиотик-индуцированные изменения микробиома могут существенно влиять на аппетит, так как уровень грелина, вызывающего аппетит, положительно коррелирует с представительством бактерий Bacteroides и Prevotella, и отрицательно – с численностью бактерий Bifidobacterium, Lactobacillus, Blautia coccoides и Eubacterium rectale. Доказано, что применение некоторых антибиотиков сопровождается не только накоплением висцерального жира, но и приводит к развитию как неалкогольной болезни печени, так и инсулинорезистентности. Рецепторы FXR и TGR5 являются сенсорами изменений микробиоты кишечника, которые участвуют в регуляции метаболических процессов макроорганизма. Развитие ожирения характеризуется наличием низкоуровневого системного воспаления. При развитии ожирения по мере увеличения размеров адипоцитов фенотип макрофагов меняется на провоспалительный фенотип М1. Накопление провоспалительных клеток в висцеральной жировой ткани является важной причиной развития инсулинорезистентности. В настоящее время необходимость применения антибиотиков при лечении инфекционных заболеваний, вызванных бактериальными агентами, не вызывает никаких клинических сомнений. Однако появление научных сведений о метаболических эффектах, возникновение которых ассоциировано с антибиотикотерапией, ставит клинические новые задачи, решение которых, вероятно, лежит в оптимизации режимов применения антибиотиков и выборе сопровождающих лекарственных средств.
This review of scientific literature is devoted to issues related to the mechanisms of antibacterial- induced adipogenesis. The antibiotics most highly associated with the development of obesity in children are the following: amoxicillin, cefotaxime, macrolides, tetracyclines, vancomycin. On the base of the results of phylogenetic, metagenomic studies of the effects of antibiotics, it was found that their use in the antenatal, early postnatal period leads to prolonged changes in both the composition and functioning of the microbiome, which is associated with the increased risk of subsequent increase of body weight of the child. The mechanisms of direct effect of antibiotics on adipogenesis are associated with their ability to increase appetite, by stimulating the release of orexin and melanin-concentrating hormone; increase the absorption of food ingredients; activate lipogenesis; induce mitochondrial dysfunction and thereby contribute to accumulation of fatty acids. The use of antibiotics significantly changes the structure of the intestinal microbiome, namely, the development of obesity is associated with a high representation of phylum bacteria Actinobacteria and Firmicutes in combination with the decrease of the number of bacteria Bacteroidetes, Verrucomicrobia and Faecalibacterium prausnitzii. Antibiotic-induced changes in the microbiome can significantly affect appetite, because the level of ghrelin that causes appetite positively correlates with the presence of bacteria Bacteroides and Prevotella, and negatively with the number of bacteria Bifidobacterium, Lactobacillus, Blautia coccoides and Eubacterium rectale. It was proved that the use of certain antibiotics is accompanied not only by the accumulation of visceral fat, but also leads to the development of both non-alcoholic liver disease and insulin resistance. The FXR and TGR5 receptors are the sensors of changes in the intestinal microbiota, which is involved in the regulation of the metabolic processes of the macroorganism. The development of obesity is characterized by the presence of low-level systemic inflammation. With the development of obesity, as the size of adipocytes increases, the macrophage phenotype changes to the pro- inflammatory M1 phenotype. The accumulation of pro-inflammatory cells in visceral adipose tissue is an important reason for development of insulin resistance. Currently, the need for antibiotics in the treatment of infectious diseases caused by bacterial agents does not raise any clinical doubts. However, the emergence of scientific information about metabolic effects, the occurrence of which is associated with antibiotic therapy, presents new clinical challenges, the solution of which probably lies in optimizing antibiotic regimens and choosing the accompanying drugs.
Collapse
|
46
|
Abstract
Microbiota is a kind of ecosystem inhabiting some organs, supporting their proper functioning, but also having a significant impact on the development of the immune system. The largest reservoir of microbiota is the digestive tract, where the largest number of lymphocytes is also present. Literature gradually increases the number of studies assessing the relationship between intestinal dysbiosis and the development of various parenteral diseases. This article presents the latest data from the medical literature regarding intestinal microbiota and barrier in patients with psoriasis. In the cited studies, a quantitative advantage of Firmicutes phylum over Bacteroidetes phylum and a smaller colonization of Actinobacteria phylum has been demonstrated. In terms of the species, colonization of bacteria Faecalibacterium prausnitzii and Akkermansia muciniphilia was reduced, and Escherichia coli increased. Regarding the participation of individual taxonomic units, the results in the cited studies are partly different. However, all revealed significant differences between the intestinal
microbiota of patients with psoriasis and a healthy population, which suggests the importance of
intestinal dysbiosis in the development of this disease. It seems more important that what leads to
disturbances in the metabolic balance is not so much the quantity of individual taxonomic units
as their disproportions. In some studies, the deviations in microbiota correlated with the level
of metabolites and indicators of inflammation. Moreover, some studies revealed a significantly
higher incidence of Candida in the oral cavity as well as in the stool samples of patients with psoriasis.
There are also reports in the literature in which the occurrence of intestinal inflammation
and the impairment of the intestinal barrier in patients with psoriasis have been demonstrated.
These observations indicate interrelations between psoriasis and intestinal disorders as well as the
involvement of dysbiosis in both associations and the pathogenesis of psoriasis.
Collapse
Affiliation(s)
- Monika Koper
- Klinika Dermatologii i Wenerologii, Uniwersytecki Szpital Kliniczny im. Wojskowej Akademii Medycznej – Centralny Szpital Weteranów w Łodzi
| | - Anna Woźniacka
- Klinika Dermatologii i Wenerologii, Uniwersytecki Szpital Kliniczny im. Wojskowej Akademii Medycznej – Centralny Szpital Weteranów w Łodzi
| | - Ewa Robak
- Klinika Dermatologii i Wenerologii, Uniwersytecki Szpital Kliniczny im. Wojskowej Akademii Medycznej – Centralny Szpital Weteranów w Łodzi
| |
Collapse
|
47
|
Sikora M, Stec A, Chrabaszcz M, Knot A, Waskiel-Burnat A, Rakowska A, Olszewska M, Rudnicka L. Gut Microbiome in Psoriasis: An Updated Review. Pathogens 2020; 9:pathogens9060463. [PMID: 32545459 PMCID: PMC7350295 DOI: 10.3390/pathogens9060463] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 05/29/2020] [Accepted: 06/08/2020] [Indexed: 12/19/2022] Open
Abstract
(1) Background: A growing body of evidence highlights that intestinal dysbiosis is associated with the development of psoriasis. The gut–skin axis is the novel concept of the interaction between skin diseases and microbiome through inflammatory mediators, metabolites and the intestinal barrier. The objective of this study was to synthesize current data on the gut microbial composition in psoriasis. (2) Methods: We conducted a systematic review of studies investigating intestinal microbiome in psoriasis, using the PRISMA checklist. We searched MEDLINE, EMBASE, and Web of Science databases for relevant published articles (2000–2020). (3) Results: All of the 10 retrieved studies reported alterations in the gut microbiome in patients with psoriasis. Eight studies assessed alpha- and beta-diversity. Four of them reported a lack of change in alpha-diversity, but all confirmed significant changes in beta-diversity. At the phylum-level, at least two or more studies reported a lower relative abundance of Bacteroidetes, and higher Firmicutes in psoriasis patients versus healthy controls. (4) Conclusions: There is a significant association between alterations in gut microbial composition and psoriasis; however, there is high heterogeneity between studies. More unified methodological standards in large-scale studies are needed to understand microbiota’s contribution to psoriasis pathogenesis and its modulation as a potential therapeutic strategy.
Collapse
Affiliation(s)
- Mariusz Sikora
- Department of Dermatology, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.C.); (A.W.-B.); (A.R.); (M.O.); (L.R.)
- Correspondence:
| | - Albert Stec
- Student Research Committee, Department of Dermatology, Medical University of Warsaw, 02-091 Warsaw, Poland; (A.S.); (A.K.)
| | - Magdalena Chrabaszcz
- Department of Dermatology, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.C.); (A.W.-B.); (A.R.); (M.O.); (L.R.)
| | - Aleksandra Knot
- Student Research Committee, Department of Dermatology, Medical University of Warsaw, 02-091 Warsaw, Poland; (A.S.); (A.K.)
| | - Anna Waskiel-Burnat
- Department of Dermatology, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.C.); (A.W.-B.); (A.R.); (M.O.); (L.R.)
| | - Adriana Rakowska
- Department of Dermatology, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.C.); (A.W.-B.); (A.R.); (M.O.); (L.R.)
| | - Malgorzata Olszewska
- Department of Dermatology, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.C.); (A.W.-B.); (A.R.); (M.O.); (L.R.)
| | - Lidia Rudnicka
- Department of Dermatology, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.C.); (A.W.-B.); (A.R.); (M.O.); (L.R.)
| |
Collapse
|
48
|
Zhang L, Wang F, Wang J, Wang Y, Fang Y. Intestinal fatty acid-binding protein mediates atherosclerotic progress through increasing intestinal inflammation and permeability. J Cell Mol Med 2020; 24:5205-5212. [PMID: 32220004 PMCID: PMC7205806 DOI: 10.1111/jcmm.15173] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 01/06/2020] [Accepted: 02/06/2020] [Indexed: 12/15/2022] Open
Abstract
Atherosclerosis is one of leading phenotypes of cardiovascular diseases, featured with increased vascular intima‐media thickness (IMT) and unstable plaques. The interaction between gastrointestinal system and cardiovascular homeostasis is emerging as a hot topic. Therefore, the present study aimed to explore the role of an intestinal protein, intestinal fatty acid‐binding protein (I‐FABP/FABP2) in the atherosclerotic progress. In western diet–fed ApoE−/− mice, FABP2 was highly expressed in intestine. Silence of intestinal Fabp2 attenuated western diet–induced atherosclerotic phenotypes, including decreasing toxic lipid accumulation, vascular fibrosis and inflammatory response. Mechanistically, intestinal Fabp2 knockdown improved intestinal permeability through increasing the expression of tight junction proteins. Meanwhile, intestinal Fabp2 knockdown mice exhibited down‐regulation of intestinal inflammation in western diet–fed ApoE−/− mice. In clinical patients, the circulating level of FABP2 was obviously increased in patients with cardiovascular disease and positively correlated with the value of carotid intima‐media thickness, total cholesterol and triglyceride. In conclusion, FABP2‐induced intestinal permeability could address a potential role of gastrointestinal system in the development of atherosclerosis, and targeting on intestinal FABP2 might provide a therapeutic approach to protect against atherosclerosis.
Collapse
Affiliation(s)
- Lulu Zhang
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| | - Fan Wang
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| | - Jiajun Wang
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| | - Yongshun Wang
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| | - Yan Fang
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| |
Collapse
|
49
|
Dowling P, Gargan S, Zweyer M, Swandulla D, Ohlendieck K. Proteomic profiling of fatty acid binding proteins in muscular dystrophy. Expert Rev Proteomics 2020; 17:137-148. [PMID: 32067530 DOI: 10.1080/14789450.2020.1732214] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: Duchenne muscular dystrophy is a neuromuscular disorder, which is caused by abnormalities in the DMD gene that encodes the membrane cytoskeletal protein dystrophin. Besides progressive skeletal muscle wasting, dystrophinopathy also affects non-skeletal muscle tissues, including cells in the cardio-respiratory system, the central nervous system, the liver and the kidney.Areas covered: This review summarizes the proteomic characterization of a key class of lipid chaperones, the large family of fatty acid binding proteins, and their potential role in muscular dystrophy. Recent proteomic surveys using animal models and patient specimens are reviewed. Pathobiochemical changes in specific proteoforms of fatty acid binding protein in the multi-system pathology of dystrophinopathy are discussed.Expert opinion: The mass spectrometric identification of distinct changes in fatty acid binding proteins in muscle, heart, liver, kidney and serum demonstrates that considerable alterations occur in key steps of metabolite transport and fat metabolism in muscular dystrophy. These new findings might be helpful to further develop a comprehensive biomarker signature of metabolic changes in X-linked muscular dystrophy, which should improve (i) our understanding of complex pathobiochemical changes due to dystrophin deficiency, (ii) the identification of novel therapeutic targets, and (iii) the design of differential diagnostic, prognostic and therapy-monitoring approaches.
Collapse
Affiliation(s)
- Paul Dowling
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland.,Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Stephen Gargan
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland.,Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Margit Zweyer
- Department of Neonatology and Pediatric Intensive Care, Children's Hospital, University of Bonn, Bonn, Germany
| | | | - Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland.,Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland
| |
Collapse
|