1
|
Rosli NFH, Mohd Nor NS, Adnan RA, Sheikh Abdul Kadir SH. A review of vitamin D deficiency and vitamin D receptor polymorphisms in endocrine-related disorders. Clin Exp Pediatr 2025; 68:30-52. [PMID: 39533737 PMCID: PMC11725620 DOI: 10.3345/cep.2024.00227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 06/05/2024] [Accepted: 07/10/2024] [Indexed: 11/16/2024] Open
Abstract
The endocrine system is a complex network of glands that produce and release hormones that regulate various physiological processes. In the past few decades, the human skin has been identified as an important peripheral endocrine organ that is the main site for the synthesis of vitamin D through exposure to sunlight. Mutations in downstream vitamin D-related gene pathways are associated with disease development. The vitamin D receptor (VDR) gene, which regulates the pleiotropic effects of vitamin D, has been extensively studied in adult populations. Several studies have reported the prevalence of vitamin D deficiency in children and adolescents. With changes in socioeconomic status and lifestyle, vitamin D-deficient individuals are prone to developing the disease at a young age. However, geographical and racial differences affect the association between VDR gene polymorphisms and vitamin D endocrine disorders, explaining the nonconsensus effects of polymorphisms and their association with disease development across populations. In this review, we discuss the connection between the vitamin D endocrine system and polymorphisms in the gene encoding VDR in children and adolescents, focusing on its effects on growth, puberty, insulin resistance, and the immune system.
Collapse
Affiliation(s)
- Nur Faten Hafizah Rosli
- Institute of Medical Molecular Biotechnology (IMMB), Faculty of Medicine, Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Sungai Buloh, Jalan Hospital, Sungai Buloh, Malaysia
| | - Noor Shafina Mohd Nor
- Institute of Medical Molecular Biotechnology (IMMB), Faculty of Medicine, Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Sungai Buloh, Jalan Hospital, Sungai Buloh, Malaysia
- Department of Paediatrics, Faculty of Medicine, Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Sungai Buloh, Jalan Hospital, Sungai Buloh, Malaysia
- Institute for Pathology, Laboratory and Forensic Medicine (I-PPerForM), Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Sungai Buloh, Jalan Hospital, Sungai Buloh, Malaysia
| | - Rose Adzrianee Adnan
- Department of Pathology, Faculty of Medicine, Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Sungai Buloh, Jalan Hospital, Sungai Buloh, Malaysia
| | - Siti Hamimah Sheikh Abdul Kadir
- Institute of Medical Molecular Biotechnology (IMMB), Faculty of Medicine, Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Sungai Buloh, Jalan Hospital, Sungai Buloh, Malaysia
- Institute for Pathology, Laboratory and Forensic Medicine (I-PPerForM), Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Sungai Buloh, Jalan Hospital, Sungai Buloh, Malaysia
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Sungai Buloh, Jalan Hospital, Sungai Buloh, Malaysia
| |
Collapse
|
2
|
Raheem MA, Rahim MA, Gul I, Reyad-Ul-Ferdous M, Zhang CY, Yu D, Pandey V, Du K, Wang R, Han S, Han Y, Qin P. COVID-19: Post infection implications in different age groups, mechanism, diagnosis, effective prevention, treatment, and recommendations. Life Sci 2024:122861. [PMID: 38925222 DOI: 10.1016/j.lfs.2024.122861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 05/28/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024]
Abstract
SARS-CoV-2 is a highly contagious pathogen that predominantly caused the COVID-19 pandemic. The persistent effects of COVID-19 are defined as an inflammatory or host response to the virus that begins four weeks after initial infection and persists for an undetermined length of time. Chronic effects are more harmful than acute ones thus, this review explored the long-term effects of the virus on various human organs, including the pulmonary, cardiovascular, and neurological, reproductive, gastrointestinal, musculoskeletal, endocrine, and lymphoid systems and found that SARS-CoV-2 adversely affects these organs of older adults. Regarding diagnosis, the RT-PCR is a gold standard method of diagnosing COVID-19; however, it requires specialized equipment and personnel for performing assays and a long time for results production. Therefore, to overcome these limitations, artificial intelligence employed in imaging and microfluidics technologies is the most promising in diagnosing COVID-19. Pharmacological and non-pharmacological strategies are the most effective treatment for reducing the persistent impacts of COVID-19 by providing immunity to post-COVID-19 patients by reducing cytokine release syndrome, improving the T cell response, and increasing the circulation of activated natural killer and CD8 T cells in blood and tissues, which ultimately reduces fever, nausea, fatigue, and muscle weakness and pain. Vaccines such as inactivated viral, live attenuated viral, protein subunit, viral vectored, mRNA, DNA, or nanoparticle vaccines significantly reduce the adverse long-term virus effects in post-COVID-19 patients; however, no vaccine was reported to provide lifetime protection against COVID-19; consequently, protective measures such as physical separation, mask use, and hand cleansing are promising strategies. This review provides a comprehensive knowledge of the persistent effects of COVID-19 on people of varying ages, as well as diagnosis, treatment, vaccination, and future preventative measures against the spread of SARS-CoV-2.
Collapse
Affiliation(s)
- Muhammad Akmal Raheem
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China
| | - Muhammad Ajwad Rahim
- College of Animal Science and Technology, Ahnui Agricultural University, Hefei, PR China
| | - Ijaz Gul
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China
| | - Md Reyad-Ul-Ferdous
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China
| | - Can Yang Zhang
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China
| | - Dongmei Yu
- School of Mechanical, Electrical & Information Engineering, Shandong University
| | - Vijay Pandey
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China
| | - Ke Du
- Department of Chemical and Environmental Engineering, University of California, Riverside, CA, USA
| | - Runming Wang
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China
| | - Sanyang Han
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China
| | - Yuxing Han
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China
| | - Peiwu Qin
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China.
| |
Collapse
|
3
|
Morin SM, Gregory KJ, Medeiros B, Terefe T, Hoshyar R, Alhusseiny A, Chen S, Schwartz RC, Jerry DJ, Vandenberg LN, Schneider SS. Benzophenone-3 exposure alters composition of tumor infiltrating immune cells and increases lung seeding of 4T1 breast cancer cells. ADVANCES IN CANCER BIOLOGY - METASTASIS 2023; 7:100080. [PMID: 37593105 PMCID: PMC10434833 DOI: 10.1016/j.adcanc.2022.100080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/19/2023]
Abstract
Environmental chemicals are a persistent and pervasive part of everyday life. A subset of environmental chemicals are xenoestrogens, compounds that bind to the estrogen receptor (ER) and drive estrogen-related processes. One such chemical, benzophenone-3 (BP3), is a common chemical in sunscreen. It is a potent UV protectant but also is quickly absorbed through the skin. While it has been approved by the FDA, there is a renewed interest in the safety of BP3, particularly in relation to breast cancer. The focus of this study was to examine the impact that BP3 has on triple negative breast cancer (TNBC) through alterations to cells in the immune microenvironment. In this study, we exposed female mice to one of two doses of BP3 before injecting them with a TNBC cell line. Several immune endpoints were examined both in the primary tissues and from in vitro studies of T cell behavior. Our studies revealed that in the lung tumor microenvironment, exposure to BP3 not only increased the number of metastases, but also the total area of tumor coverage. We also found that BP3 caused alterations in immune populations in a tissue-dependent manner, particularly in T cells. Taken together, our data suggest that while BP3 may not directly affect the proliferation of TNBC, growth and metastasis of TNBC-derived tumors can be altered by BP3 exposures via the alterations in the immune populations of the tumor microenvironment.
Collapse
Affiliation(s)
- Stephanie M. Morin
- Pioneer Valley Life Sciences Institute, Springfield, MA, 01199, USA
- Dept of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, 01003, USA
| | - Kelly J. Gregory
- Pioneer Valley Life Sciences Institute, Springfield, MA, 01199, USA
| | - Brenda Medeiros
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts, Amherst, 01003, USA
| | - Tigist Terefe
- Pioneer Valley Life Sciences Institute, Springfield, MA, 01199, USA
| | - Reyhane Hoshyar
- Breast Cancer and the Environment Research Program, Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | - Ahmed Alhusseiny
- University of Massachusetts Chan Medical School-Baystate, Department of Pathology, Springfield, MA, 01199, USA
| | - Shiuan Chen
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Richard C. Schwartz
- Breast Cancer and the Environment Research Program, Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | - D. Joseph Jerry
- Dept of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, 01003, USA
| | - Laura N. Vandenberg
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts, Amherst, 01003, USA
| | - Sallie S. Schneider
- Pioneer Valley Life Sciences Institute, Springfield, MA, 01199, USA
- Dept of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, 01003, USA
- University of Massachusetts Chan Medical School-Baystate, Department of Surgery, Springfield, MA, 01199, USA
| |
Collapse
|
4
|
Bera AK, Chowdhury H, Ghatak S, Malick RC, Chakraborty N, Chakraborty HJ, Swain HS, Hassan MA, Das BK. Microbiome analysis reveals potential for modulation of gut microbiota through polysaccharide-based prebiotic feeding in Oreochromis niloticus (Linnaeus, 1758). Front Physiol 2023; 14:1168284. [PMID: 37362433 PMCID: PMC10285058 DOI: 10.3389/fphys.2023.1168284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 05/23/2023] [Indexed: 06/28/2023] Open
Abstract
Characterization and functional profiling of the gut microbiota are essential for guiding nutritional interventions in fish and achieving favorable host-microbe interactions. Thus, we conducted a 30 days study to explore and document the gut microbial community of O. niloticus, as well as to evaluate the effects of a polysaccharide-based prebiotics with 0.5% and 0.75% Aloe vera extract on the gut microbiome through genomic analysis. The V3-V4 region of 16S rRNA was amplified and sequenced using Illumina HiSeq 2500, resulting in 1,000,199 reads for operational taxonomic unit (OTU) identification. Out of 8,894 OTUs, 1,181 were selected for further analysis. Our results revealed that Planctomycetes, Firmicutes, Proteobacteria, Verrucomicrobia, Actinobacteria, and Fusobacteria were the dominant phyla in both control and treatment samples. Higher doses of prebiotics were found to improve Planctomycetes and Firmicutes while decreasing Proteobacteria and Verrucomicrobia. We observed increasing trends in the abundance of Bacilli, Bacillaceae, and Bacillus bacteria at the class, family, and genus levels, respectively, in a dose-dependent manner. These findings were consistent with the conventional colony count data, which showed a higher prevalence of Bacillus in prebiotic-supplemented groups. Moreover, predicted functional analysis using PICRUSt indicated a dose-dependent upregulation in glycolysis V, superpathway of glycol metabolism and degradation, glucose and xylose degradation, glycolysis II, and sulfoglycolysis pathways. Most of the energy, protein, and amino acid synthesis pathways were upregulated only at lower doses of prebiotic treatment. Our findings suggest that the gut microbiome of O. niloticus can be optimized through nutritional interventions with plant-based polysaccharides for improved growth performance in commercial fish.
Collapse
Affiliation(s)
- Asit Kumar Bera
- Central Inland Fisheries Research Institute (ICAR), Bārākpur, India
| | | | - Sandeep Ghatak
- The ICAR Research Complex for North Eastern Hill Region (ICAR RC NEH), Umiam, India
| | | | | | | | | | - M. A. Hassan
- Central Inland Fisheries Research Institute (ICAR), Bārākpur, India
| | | |
Collapse
|
5
|
Borzyszkowska D, Niedzielska M, Kozłowski M, Brodowska A, Przepiera A, Malczyk-Matysiak K, Cymbaluk-Płoska A, Sowińska-Przepiera E. Evaluation of Hormonal Factors in Acne Vulgaris and the Course of Acne Vulgaris Treatment with Contraceptive-Based Therapies in Young Adult Women. Cells 2022; 11:4078. [PMID: 36552842 PMCID: PMC9777314 DOI: 10.3390/cells11244078] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Acne vulgaris is a common chronic inflammatory skin disease, which is considered one of the diseases of civilization due to the significant influence of environmental factors on the severity and frequency of these lesions. The aim of this study was to evaluate the hormonal profile of patients before treatment and to assess selected hormonal parameters after treatment. Our first objective was to examine the correlation between the selected hormonal parameters and the severity of acne before treatment. Our second objective was to evaluate the impact of treatment with three therapies, as measured by the selected hormonal parameters and acne severity. Statistical calculations were performed using the R v.4.1.1 statistical calculation environment (IDE RStudio v. 1.4.1717) with a significance level for the statistical tests set at α = 0.05. The results showed that the women in the pre-treatment (T1) and control (C) groups had significant differences in testosterone, androstendione, FAI, SHBG, prolactin, ACTH, and cortisol concentrations. After treatment, there were still significant differences in testosterone, androstendione, FAI, and SHBG concentrations between the post-treatment (T2) and control groups. We concluded that testosterone, androstendione, and cortisol concentrations correlate with acne severity. Acne in adult women may be an important clinical marker of androgen excess syndrome and cannot be considered a transient symptom of puberty. The mainstay of acne treatment is contraceptive therapy (ethonylestradiol and drospirenone). In this study, we confirmed the effectiveness of three contraceptive-based treatments using hormonal parameters and acne severity.
Collapse
Affiliation(s)
- Dominika Borzyszkowska
- Department of Reconstructive Surgery and Gynecological Oncology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
| | - Mirela Niedzielska
- Department of Endocrinology, Metabolic and Internal Diseases, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, 71-252 Szczecin, Poland
| | - Mateusz Kozłowski
- Department of Reconstructive Surgery and Gynecological Oncology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
| | - Agnieszka Brodowska
- Department of Gynecology, Endocrinology and Gynecological Oncology, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, 71-252 Szczecin, Poland
| | - Adam Przepiera
- Department of Urology and Urologic Oncology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
| | - Kinga Malczyk-Matysiak
- Department of Endocrinology, Metabolic and Internal Diseases, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, 71-252 Szczecin, Poland
| | - Aneta Cymbaluk-Płoska
- Department of Reconstructive Surgery and Gynecological Oncology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
| | - Elżbieta Sowińska-Przepiera
- Department of Endocrinology, Metabolic and Internal Diseases, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, 71-252 Szczecin, Poland
- Pediatric, Adolescent Gynecology Clinic, Department of Gynecology, Endocrinology and Gynecological Oncology, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, 71-252 Szczecin, Poland
| |
Collapse
|
6
|
Gruszecka‐Kosowska A, Ampatzoglou A, Aguilera‐Gómez M. Microbiota analysis for risk assessment of xenobiotics: cumulative xenobiotic exposure and impact on human gut microbiota under One Health approach. EFSA J 2022; 20:e200916. [PMID: 36531282 PMCID: PMC9749440 DOI: 10.2903/j.efsa.2022.e200916] [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: 12/15/2022] Open
Abstract
Human gut microbiota is the microbial community that, through the constant bidirectional communication with its host, plays the critical role of maintaining the state of eubiosis and health balance, contributing to food digestion, detoxification, and proper endocrine, neurological, immunological and potentially reproductive health. To this extent, gut microbiota is called the 'second brain' as well as the 'second liver'. Xenobiotics, including environmental pollutants, are widely spread in the environment and easily accessible in food, cosmetics, personal care products, drugs and medicinal products. Thus, the gut microbiota can be exposed to these xenobiotics, which in turn might alter its composition and metabolism that can trigger dysbiosis, and they seem associated with disorders and diseases in the host. A specific group of xenobiotics, called endocrine-disrupting chemicals, is particularly important due to relevant adverse health effects. A considerable challenge in risk assessment is the combined exposure to xenobiotics, for which the integrated approaches, including the One Health concept, are still under development. Nevertheless, recent research advancements focus on molecular data in the search for elucidating crucial microbiome biomarkers, associated with physiopathology and specific dysfunctions triggered by xenobiotic exposure. In this context, the application of meta-omics and integration of genomics, metagenomics, metabolomics, metatranscriptomics, proteomics and multidisciplinary approaches are particularly important.
Collapse
Affiliation(s)
| | - Antonios Ampatzoglou
- University of Granada (INYTA‐UGR)Institute of Nutrition and Food TechnologyGranadaSpain
| | | |
Collapse
|
7
|
Kowalczyk A, Wrzecińska M, Czerniawska-Piątkowska E, Araújo JP, Cwynar P. Molecular consequences of the exposure to toxic substances for the endocrine system of females. Biomed Pharmacother 2022; 155:113730. [PMID: 36152416 DOI: 10.1016/j.biopha.2022.113730] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/05/2022] [Accepted: 09/19/2022] [Indexed: 11/02/2022] Open
Abstract
Endocrine-disrupting chemicals (EDCs) are common in the environment and in everyday products such as cosmetics, plastic food packaging, and medicines. These substances are toxic in small doses (even in the order of micrograms) and enter the body through the skin, digestive or respiratory system. Numerous studies confirm the negative impact of EDCs on living organisms. They disrupt endocrine functions, contributing to the development of neoplastic and neurological diseases, as well as problems with the circulatory system and reproduction. EDCs affect humans and animals by modulating epigenetic processes that can lead to disturbances in gene expression or failure and even death. They also affect steroid hormones by binding to their receptors as well as interfering with synthesis and secretion of hormones. Prenatal exposure may be related to the impact of EDCs on offspring, resulting in effects of these substances on the ovaries and leading to the reduction of fertility through disturbances in the function of steroid receptors or problems with steroidogenesis and gametogenesis. Current literature indicates the need to continue research on the effects of EDCs on the female reproductive system. The aim of this review was to identify the effects of endocrine-disrupting chemicals on the female reproductive system and their genetic effects based on recent literature.
Collapse
Affiliation(s)
- Alicja Kowalczyk
- Department of Environmental Hygiene and Animal Welfare, Wrocław University of Environmental and Life Sciences, Chełmońskiego 38C, Wrocław, Poland.
| | - Marcjanna Wrzecińska
- Department of Ruminant Science, West Pomeranian University of Technology, Klemensa Janickiego 29, 71-270 Szczecin, Poland.
| | - Ewa Czerniawska-Piątkowska
- Department of Ruminant Science, West Pomeranian University of Technology, Klemensa Janickiego 29, 71-270 Szczecin, Poland.
| | - José Pedro Araújo
- Mountain Research Centre (CIMO), Instituto Politécnico de Viana do Castelo, Rua D. Mendo Afonso, 147, Refóios do Lima, 4990-706 Ponte de Lima, Portugal.
| | - Przemysław Cwynar
- Department of Environmental Hygiene and Animal Welfare, Wrocław University of Environmental and Life Sciences, Chełmońskiego 38C, Wrocław, Poland.
| |
Collapse
|
8
|
Wang Z, Tian X, Wang C, Qi X, Gracia‐Sancho J, Dong L. Transforming one organ into another to overcome challenges in tissue engineering. PORTAL HYPERTENSION & CIRRHOSIS 2022; 1:116-124. [DOI: 10.1002/poh2.26] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 07/11/2022] [Indexed: 01/03/2025]
Abstract
AbstractTissue engineering (TE) is promising for the regeneration of failed organs. However, immune rejection, shortage of seed cells, and unintegrated blood vessels restrict the development and clinical application of TE. The last factor is the most challenging and intractable. Harnessing the mature blood vessel network in existing dispensable organs could be a powerful approach to effectively overcome the obstacles. After being remodeled to harbor an immunosuppressive and proregenerative niche, these potential target organs can be transformed into other organs with specific physiological functions, compensating the latter's failed native functions. Organ transformation, such as a hepatized spleen, represents an effective and encouraging TE strategy. In this review, we discuss the current development and obstacles of TE and its feasibility and superiority in organ transformation.
Collapse
Affiliation(s)
- Zhenzhen Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences Nanjing University Nanjing Jiangsu China
| | - Xuejiao Tian
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences Nanjing University Nanjing Jiangsu China
| | - Chunming Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences University of Macau Taipa Macau SAR China
| | - Xiaolong Qi
- CHESS Center, Institute of Portal Hypertension The First Hospital of Lanzhou University Lanzhou Gansu China
| | - Jordi Gracia‐Sancho
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital University of Bern Bern Switzerland
- Liver Vascular Biology Research Group IDIBAPS Research Institute, CIBEREHD Barcelona Spain
| | - Lei Dong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences Nanjing University Nanjing Jiangsu China
- Chemistry and Biomedicine Innovative Center Nanjing University Nanjing Jiangsu China
| |
Collapse
|
9
|
Cusick JA, Wellman CL, Demas GE. Maternal stress and the maternal microbiome have sex-specific effects on offspring development and aggressive behavior in Siberian hamsters (Phodopus sungorus). Horm Behav 2022; 141:105146. [PMID: 35276524 DOI: 10.1016/j.yhbeh.2022.105146] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 11/29/2022]
Abstract
The gut microbiome, a community of commensal, symbiotic and pathogenic bacteria, fungi, and viruses, interacts with many physiological systems to affect behavior. Prenatal experiences, including exposure to maternal stress and different maternal microbiomes, are important sources of organismal variation that can affect offspring development. These physiological systems do not act in isolation and can have long-term effects on offspring development and behavior. Here we investigated the interactive effects of maternal stress and manipulations of the maternal microbiome on offspring development and social behavior using Siberian hamsters, Phodopus sungorus. We exposed pregnant females to either a social stressor, antibiotics, both the social stressor and antibiotics, or no treatment (i.e., control) over the duration of their pregnancy and quantified male and female offspring growth, gut microbiome composition and diversity, stress-induced cortisol concentrations, and social behavior. Maternal antibiotic exposure altered the gut microbial communities of male and female offspring. Maternal treatment also had sex-specific effects on aspects of offspring development and aggressive behavior. Female offspring produced by stressed mothers were more aggressive than other female offspring. Female, but not male, offspring produced by mothers exposed to the combined treatment displayed low levels of aggression, suggesting that alteration of the maternal microbiome attenuated the effects of prenatal stress in a sex-specific manner. Maternal treatment did not affect non-aggressive behavior in offspring. Collectively, our study offers insight into how maternal systems can interact to affect offspring in sex-specific ways and highlights the important role of the maternal microbiome in mediating offspring development and behavior.
Collapse
Affiliation(s)
- Jessica A Cusick
- Department of Biology, Utah Valley University, United States of America; Department of Biology, Indiana University, United States of America; Animal Behavior Program, Indiana University, United States of America.
| | - Cara L Wellman
- Animal Behavior Program, Indiana University, United States of America; Department of Psychological and Brain Sciences, Indiana University, United States of America; Program in Neuroscience, Indiana University, United States of America
| | - Gregory E Demas
- Department of Biology, Indiana University, United States of America; Animal Behavior Program, Indiana University, United States of America; Program in Neuroscience, Indiana University, United States of America
| |
Collapse
|
10
|
Lv W, Jin S, Cao D, Wang N, Jin X, Zhang Y. Effects of Luteinizing Hormone Releasing Hormone A2 on Gonad Development in Juvenile Amur Sturgeon, Acipenser schrenckii, Revealed by Transcriptome Profiling Analysis. Front Genet 2022; 13:859965. [PMID: 35401695 PMCID: PMC8989137 DOI: 10.3389/fgene.2022.859965] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 02/22/2022] [Indexed: 11/15/2022] Open
Abstract
Acipenser schrenckii is an economically important aquatic species whose gonads require particularly long times to reach sexual maturity. Luteinizing hormone plays important roles in gonad development, and luteinizing hormone releasing hormone A2 (LH-A2) is used as an oxytocin to promote ovulation in aquaculture of A. schrenckii. In this study, we aimed to determine the effects of LH-A2 on gonad development in juvenile A. schrenckii through transcriptome profiling analysis of the pituitary and gonads after LH-A2 treatment at a dose of 3 μg/kg. The 17β-estradiol (E2) levels gradually increased with LH-A2 treatment time, and significantly differed from those of the control group on days 5 and 7 (p < 0.01). However, the content of testosterone (Testo) gradually decreased with LH-A2 treatment time and showed significant differences on day 3 (p < 0.05), and on days 5 and 7 (p < 0.01), compared to those in the control group. Thus, LH-A2 promotes the secretion of E2 and inhibits the secretion of Testo. Transcriptome profiling analysis revealed a total of 2,883 and 8,476 differentially expressed genes (DEGs) in the pituitary and gonads, respectively, thus indicating that LH-A2 has more regulatory effects on the gonads than the pituitary in A. schrenckii. Signal transduction, global and overview maps, immune system, endocrine system and lipid metabolism were the main enriched metabolic pathways in both the pituitary and gonads. Sixteen important genes were selected from these metabolic pathways. Seven genes were co-DEGs enriched in both signal transduction and endocrine system metabolic pathways. The other co-DEGs were selected from the immune system and lipid metabolism metabolic pathways, and showed mRNA expression changes of >7.0. The expression of five DEGs throughout LH-A2 treatment was verified to show the same patterns of change as those observed with RNA-seq, indicating the accuracy of the RNA-seq in this study. Our findings provide valuable evidence of the regulation of gonad development of juvenile A. schrenckii by LH-A2 and may enable the establishment of artificial techniques to regulate gonad development in this species.
Collapse
Affiliation(s)
- Weihua Lv
- Key Open Laboratory of Cold Water Fish Germplasm Resources and Breeding of Heilongjiang Province, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, China
| | - Shubo Jin
- Key Open Laboratory of Cold Water Fish Germplasm Resources and Breeding of Heilongjiang Province, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, China
| | - Dingchen Cao
- Key Open Laboratory of Cold Water Fish Germplasm Resources and Breeding of Heilongjiang Province, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, China
| | - Nianmin Wang
- Key Open Laboratory of Cold Water Fish Germplasm Resources and Breeding of Heilongjiang Province, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, China
| | - Xing Jin
- Key Open Laboratory of Cold Water Fish Germplasm Resources and Breeding of Heilongjiang Province, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, China
| | - Ying Zhang
- Key Open Laboratory of Cold Water Fish Germplasm Resources and Breeding of Heilongjiang Province, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, China
| |
Collapse
|
11
|
Gratton R, Del Vecchio C, Zupin L, Crovella S. Unraveling the Role of Sex Hormones on Keratinocyte Functions in Human Inflammatory Skin Diseases. Int J Mol Sci 2022; 23:3132. [PMID: 35328552 PMCID: PMC8955788 DOI: 10.3390/ijms23063132] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/09/2022] [Accepted: 03/11/2022] [Indexed: 12/23/2022] Open
Abstract
The skin exerts several fundamental functions that are the first physical, chemical and immune barriers to the human body. Keratinocytes, the main cell type of the epidermis, provide mechanical defense, support skin integrity and actively endorse cutaneous immune responses. Not surprisingly, considering these crucial activities, alterations in keratinocyte functions are associated with different inflammatory skin diseases. Recent findings indicate that the skin should not only be regarded as a target for hormones but that it should also be considered as an endocrine peripheral organ that is directly involved in the synthesis and metabolism of these chemical messengers. Sex hormones have multiple effects on the skin, attributed to the binding with intracellular receptors expressed by different skin cell populations, including keratinocytes, that activate downstream signaling routes that modulate specific cellular functions and activities. This review is aimed at reorganizing the current knowledge on the role exerted by sex hormones on keratinocyte function in five different inflammatory skin diseases: Hidradenitis suppurativa; Acne vulgaris; Atopic dermatitis; progesterone hypersensitivity; psoriasis. The results of our work aim to provide a deeper insight into common cellular mechanisms and molecular effectors that might constitute putative targets to address for the development of specific therapeutic interventions.
Collapse
Affiliation(s)
- Rossella Gratton
- Department of Advanced Diagnostics, Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 34137 Trieste, Italy
| | - Cecilia Del Vecchio
- Dermatology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Luisa Zupin
- Maternal-Neonatal Department, Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 34137 Trieste, Italy;
| | - Sergio Crovella
- Biological Sciences Program, Department of Biological and Environmental Sciences, College of Arts and Sciences, University of Qatar, Doha 2713, Qatar;
| |
Collapse
|
12
|
Yang Y, Wang Y. Autocrine, Paracrine, and Endocrine Signals That Can Alter Alveolar Macrophages Function. Rev Physiol Biochem Pharmacol 2022; 186:177-198. [PMID: 36472676 DOI: 10.1007/112_2022_76] [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] [Indexed: 12/12/2022]
Abstract
Alveolar macrophages (AMs) are extremely versatile cells with complex functions involved in health or diseases such as pneumonia, asthma, and pulmonary alveolar proteinosis. In recent years, it has been widely identified that the different functions and states of macrophages are the results from the complex interplay between microenvironmental signals and macrophage lineage. Diverse and complicated signals to which AMs respond are mentioned when they are described individually or in a particular state of AMs. In this review, the microenvironmental signals are divided into autocrine, paracrine, and endocrine signals based on their secreting characteristics. This new perspective on classification provides a more comprehensive and systematic introduction to the complex signals around AMs and is helpful for understanding the roles of AMs affected by physiological environment. The existing possible treatments of AMs are also mentioned in it. The thorough understanding of AMs signals modulation may be contributed to the development of more effective therapies for AMs-related lung diseases.
Collapse
Affiliation(s)
- Yue Yang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yun Wang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, People's Republic of China.
| |
Collapse
|
13
|
Keady MM, Prado N, Lim HC, Brown J, Paris S, Muletz-Wolz CR. Clinical health issues, reproductive hormones, and metabolic hormones associated with gut microbiome structure in African and Asian elephants. Anim Microbiome 2021; 3:85. [PMID: 34930501 PMCID: PMC8686393 DOI: 10.1186/s42523-021-00146-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 11/25/2021] [Indexed: 12/26/2022] Open
Abstract
Background The gut microbiome is important to immune health, metabolism, and hormone regulation. Understanding host–microbiome relationships in captive animals may lead to mediating long term health issues common in captive animals. For instance, zoo managed African elephants (Loxodonta africana) and Asian elephants (Elephas maximus) experience low reproductive rates, high body condition, and gastrointestinal (GI) issues. We leveraged an extensive collection of fecal samples and health records from the Elephant Welfare Study conducted across North American zoos in 2012 to examine the link between gut microbiota and clinical health issues, reproductive hormones, and metabolic hormones in captive elephants. We quantified gut microbiomes of 69 African and 48 Asian elephants from across 50 zoos using Illumina sequencing of the 16S rRNA bacterial gene.
Results Elephant species differed in microbiome structure, with African elephants having lower bacterial richness and dissimilar bacterial composition from Asian elephants. In both species, bacterial composition was strongly influenced by zoo facility. Bacterial richness was lower in African elephants with recent GI issues, and richness was positively correlated with metabolic hormone total triiodothyronine (total T3) in Asian elephants. We found species-specific associations between gut microbiome composition and hormones: Asian elephant gut microbiome composition was linked to total T3 and free thyroxine (free T4), while fecal glucocorticoid metabolites (FGM) were linked to African elephant gut microbiome composition. We identified many relationships between bacterial relative abundances and hormone concentrations, including Prevotella spp., Treponema spp., and Akkermansia spp.
Conclusions We present a comprehensive assessment of relationships between the gut microbiome, host species, environment, clinical health issues, and the endocrine system in captive elephants. Our results highlight the combined significance of host species-specific regulation and environmental effects on the gut microbiome between two elephant species and across 50 zoo facilities. We provide evidence of clinical health issues, reproductive hormones, and metabolic hormones associated with the gut microbiome structure of captive elephants. Our findings establish the groundwork for future studies to investigate bacterial function or develop tools (e.g., prebiotics, probiotics, dietary manipulations) suitable for conservation and zoo management. Supplementary Information The online version contains supplementary material available at 10.1186/s42523-021-00146-9.
Collapse
Affiliation(s)
- Mia M Keady
- School for Systems Biology, George Mason University, Fairfax, VA, USA. .,Center for Conservation Genomics, Smithsonian National Zoo & Conservation Biology Institute, Washington, DC, USA.
| | - Natalia Prado
- School for Systems Biology, George Mason University, Fairfax, VA, USA. .,Center for Conservation Genomics, Smithsonian National Zoo & Conservation Biology Institute, Washington, DC, USA. .,Center for Species Survival, Smithsonian National Zoo & Conservation Biology Institute, Front Royal, VA, USA. .,Department of Biology, Adelphi University, Garden City, NY, USA.
| | - Haw Chuan Lim
- School for Systems Biology, George Mason University, Fairfax, VA, USA.,Center for Conservation Genomics, Smithsonian National Zoo & Conservation Biology Institute, Washington, DC, USA
| | - Janine Brown
- Center for Species Survival, Smithsonian National Zoo & Conservation Biology Institute, Front Royal, VA, USA
| | - Steve Paris
- Center for Species Survival, Smithsonian National Zoo & Conservation Biology Institute, Front Royal, VA, USA
| | - Carly R Muletz-Wolz
- Center for Conservation Genomics, Smithsonian National Zoo & Conservation Biology Institute, Washington, DC, USA.
| |
Collapse
|
14
|
Cusick JA, Wellman CL, Demas GE. The call of the wild: using non-model systems to investigate microbiome-behaviour relationships. J Exp Biol 2021; 224:jeb224485. [PMID: 33988717 PMCID: PMC8180253 DOI: 10.1242/jeb.224485] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
On and within most sites across an animal's body live complex communities of microorganisms. These microorganisms perform a variety of important functions for their hosts, including communicating with the brain, immune system and endocrine axes to mediate physiological processes and affect individual behaviour. Microbiome research has primarily focused on the functions of the microbiome within the gastrointestinal tract (gut microbiome) using biomedically relevant laboratory species (i.e. model organisms). These studies have identified important connections between the gut microbiome and host immune, neuroendocrine and nervous systems, as well as how these connections, in turn, influence host behaviour and health. Recently, the field has expanded beyond traditional model systems as it has become apparent that the microbiome can drive differences in behaviour and diet, play a fundamental role in host fitness and influence community-scale dynamics in wild populations. In this Review, we highlight the value of conducting hypothesis-driven research in non-model organisms and the benefits of a comparative approach that assesses patterns across different species or taxa. Using social behaviour as an intellectual framework, we review the bidirectional relationship between the gut microbiome and host behaviour, and identify understudied mechanisms by which these effects may be mediated.
Collapse
Affiliation(s)
- Jessica A. Cusick
- Department of Biology, Indiana University, Biology Building 142, 1001 East Third Street, Bloomington, IN 47405, USA
- Animal Behavior Program, Indiana University, 409 N. Park Avenue, Bloomington, IN 47405, USA
| | - Cara L. Wellman
- Animal Behavior Program, Indiana University, 409 N. Park Avenue, Bloomington, IN 47405, USA
- Department of Psychological and Brain Sciences, Indiana University, 1101 E. 10th Street, Bloomington, IN 47405-7007, USA
- Program in Neuroscience, Indiana University, Psychology Building, 1101 E 10th Street Bloomington, IN 47405-2204, USA
| | - Gregory E. Demas
- Department of Biology, Indiana University, Biology Building 142, 1001 East Third Street, Bloomington, IN 47405, USA
- Animal Behavior Program, Indiana University, 409 N. Park Avenue, Bloomington, IN 47405, USA
- Program in Neuroscience, Indiana University, Psychology Building, 1101 E 10th Street Bloomington, IN 47405-2204, USA
| |
Collapse
|
15
|
Ghareghani M, Ghanbari A, Eid A, Shaito A, Mohamed W, Mondello S, Zibara K. Hormones in experimental autoimmune encephalomyelitis (EAE) animal models. Transl Neurosci 2021; 12:164-189. [PMID: 34046214 PMCID: PMC8134801 DOI: 10.1515/tnsci-2020-0169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 04/05/2021] [Accepted: 04/14/2021] [Indexed: 12/30/2022] Open
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS) in which activated immune cells attack the CNS and cause inflammation and demyelination. While the etiology of MS is still largely unknown, the interaction between hormones and the immune system plays a role in disease progression, but the mechanisms by which this occurs are incompletely understood. Several in vitro and in vivo experimental, but also clinical studies, have addressed the possible role of the endocrine system in susceptibility and severity of autoimmune diseases. Although there are several demyelinating models, experimental autoimmune encephalomyelitis (EAE) is the oldest and most commonly used model for MS in laboratory animals which enables researchers to translate their findings from EAE into human. Evidences imply that there is great heterogeneity in the susceptibility to the induction, the method of induction, and the response to various immunological or pharmacological interventions, which led to conflicting results on the role of specific hormones in the EAE model. In this review, we address the role of endocrine system in EAE model to provide a comprehensive view and a better understanding of the interactions between the endocrine and the immune systems in various models of EAE, to open up a ground for further detailed studies in this field by considering and comparing the results and models used in previous studies.
Collapse
Affiliation(s)
- Majid Ghareghani
- Neuroscience Laboratory, CHU de Québec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec City, QC, Canada
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Amir Ghanbari
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Ali Eid
- Biomedical and Pharmaceutical Research Unit and Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Abdullah Shaito
- Department of Biological and Chemical Sciences, Faculty of Arts and Sciences, Lebanese International University, Beirut, Lebanon
| | - Wael Mohamed
- Clinical Pharmacology Department, Menoufia Medical School, Menoufia University, Shibin Al Kawm, Egypt
- Department of Basic Medical Sciences, Kulliyyah of Medicine, International Islamic University Malaysia (IIUM), Kuantan, Pahang, Malaysia
| | - Stefania Mondello
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Kazem Zibara
- PRASE, Lebanese University, Beirut, Lebanon
- Biology Department, Faculty of Sciences – I, Lebanese University, Beirut, Lebanon
| |
Collapse
|
16
|
Lv W, Jin S, Wang N, Cao D, Jin X, Zhang Y. Identification of important proteins from the gonads and pituitary involved in the gonad development of Amur sturgeon, Acipenser schrenckii, regulated by GnRH-a treatment by iTRAQ-based analysis. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2021; 39:100831. [PMID: 33933834 DOI: 10.1016/j.cbd.2021.100831] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 02/16/2021] [Accepted: 03/26/2021] [Indexed: 11/16/2022]
Affiliation(s)
- Weihua Lv
- Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Haebin, China
| | - Shubo Jin
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China
| | - Nianmin Wang
- Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Haebin, China
| | - Dingchen Cao
- Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Haebin, China
| | - Xing Jin
- Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Haebin, China.
| | - Ying Zhang
- Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Haebin, China.
| |
Collapse
|
17
|
Williams CL, Garcia-Reyero N, Martyniuk CJ, Tubbs CW, Bisesi JH. Regulation of endocrine systems by the microbiome: Perspectives from comparative animal models. Gen Comp Endocrinol 2020; 292:113437. [PMID: 32061639 DOI: 10.1016/j.ygcen.2020.113437] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 01/29/2020] [Accepted: 02/10/2020] [Indexed: 02/07/2023]
Abstract
The microbiome regulates endocrine systems and influences many aspects of hormone signaling. Using examples from different animal taxa, we highlight the state of the science in microbiome research as it relates to endocrinology and endocrine disruption research. Using a comparative approach discussing fish, birds, and mammals, we demonstrate the bidirectional interaction between microbiota and hormone systems, presenting concepts that include (1) gastrointestinal microbiome regulation of the neuroendocrine feeding axis; (2) stress hormones and microbial communities; (3) the role of site-specific microbiota in animal reproduction; (4) microbiome effects on the neuroendocrine systems and behavior; and (5) novel mechanisms of endocrine disruption through the microbiome. This mini-review demonstrates that hormones can directly affect the richness and diversity of microbiota and conversely, microbiota can influence hormone production and mediate their functions in animals. In addition, microbiota can influence the action of a diverse range of neurotransmitters and neuropeptides in the central nervous system, which can lead to behavioral disruptions. As many animals have species-specific reproductive behaviors, it is important to understand how shifts in the microbiota relate to these complex interactions between sexes. This is especially important for captive animals on specialized diets, and there are significant implications for microbiome research in conservation and reproductive biology. For example, microbial metabolites may modify motility of gametes or modulate hormone-receptor interactions in reproductive tissues. Thus, efforts to incorporate metabolomics into the science of microbiome-endocrine relationships, both those produced by the host and those generated from microbial metabolism, are increasingly needed. These concepts have fostered an exciting emerging era in comparative endocrinology.
Collapse
Affiliation(s)
- Candace L Williams
- Reproductive Sciences, San Diego Zoo Global Institute for Conservation Research, Escondido, CA 92027, USA.
| | - Natàlia Garcia-Reyero
- Environmental Laboratory, US Army Engineer Research & Development Center, Vicksburg, MS 39180, USA
| | - Christopher J Martyniuk
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida Genetics Institute, Interdisciplinary Program in Biomedical Sciences Neuroscience, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Christopher W Tubbs
- Reproductive Sciences, San Diego Zoo Global Institute for Conservation Research, Escondido, CA 92027, USA
| | - Joseph H Bisesi
- Department of Environmental and Global Health and Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
18
|
Butt RL, Volkoff H. Gut Microbiota and Energy Homeostasis in Fish. Front Endocrinol (Lausanne) 2019; 10:9. [PMID: 30733706 PMCID: PMC6353785 DOI: 10.3389/fendo.2019.00009] [Citation(s) in RCA: 184] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 01/09/2019] [Indexed: 12/25/2022] Open
Abstract
The microorganisms within the intestinal tract (termed gut microbiota) have been shown to interact with the gut-brain axis, a bidirectional communication system between the gut and the brain mediated by hormonal, immune, and neural signals. Through these interactions, the microbiota might affect behaviors, including feeding behavior, digestive/absorptive processes (e.g., by modulating intestinal motility and the intestinal barrier), metabolism, as well as the immune response, with repercussions on the energy homeostasis and health of the host. To date, research in this field has mostly focused on mammals. Studies on non-mammalian models such as fish may provide novel insights into the specific mechanisms involved in the microbiota-brain-gut axis. This review describes our current knowledge on the possible effects of microbiota on feeding, digestive processes, growth, and energy homeostasis in fish, with emphasis on the influence of brain and gut hormones, environmental factors, and inter-specific differences.
Collapse
Affiliation(s)
| | - Helene Volkoff
- Departments of Biology and Biochemistry, Memorial University of Newfoundland, St. John's, NL, Canada
| |
Collapse
|
19
|
Li J, Zhang Y, Chen L, Lu X, Li Z, Xue Y, Guan YQ. Cervical Cancer HeLa Cell Autocrine Apoptosis Induced by Coimmobilized IFN-γ plus TNF-α Biomaterials. ACS APPLIED MATERIALS & INTERFACES 2018; 10:8451-8464. [PMID: 29436216 DOI: 10.1021/acsami.7b18277] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Using external methods to induce the death of cancer cells is recognized as one of the main strategies for cancer treatment. Research indicated that TNF-α is frequently used in tumor biotherapy while IFN-γ can directly inhibit tumor cell proliferation. In our study, TNF-α and IFN-γ were coimmobilized on polystyrene material (PSt) or Fe3O4-oleic acid nanoparticles (NPs). Then the structural change of these two proteins can be observed. Meanwhile, the expressions of both TNF-α and IFN-α increased significantly, as determined by gene microarray analysis; however, in the presence of TNF-α plus IFN-α inhibitors, TNF-α and IFN-α did not increase in HeLa cells induced by coimmobilized IFN-γ plus TNF-α. Our results indicate that such change can stimilate HeLa cells to secrete more TNF-α and IFN-α, by which the apoptosis of HeLa cells could be further induced. This study is the first report of autocrine-induced apoptosis of HeLa cells. In addition, we performed ELISA, RT-PCR, flow cytometry, and Western blot analyses, as well as a series of analytical tests at the animal level. our data also indicate that the PSt-coimmobilized IFN-γ plus TNF-α has apparent effects for cancer treatment in vivo, which is of great significance for translation into clinical medicine.
Collapse
Affiliation(s)
- Jian Li
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics , South China Normal University , Guangzhou 510631 , China
- Joint Laboratory of Laser Oncology with Cancer Center of Sun Yet-sen University , South China Normal University , Guangzhou 510631 , China
| | - Yuxiao Zhang
- School of Life Science , South China Normal University , Guangzhou 510631 , China
| | - Liyi Chen
- School of Life Science , South China Normal University , Guangzhou 510631 , China
| | - Xinhua Lu
- School of Life Science , South China Normal University , Guangzhou 510631 , China
| | - Zhibin Li
- School of Life Science , South China Normal University , Guangzhou 510631 , China
| | - Yongyong Xue
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics , South China Normal University , Guangzhou 510631 , China
- Joint Laboratory of Laser Oncology with Cancer Center of Sun Yet-sen University , South China Normal University , Guangzhou 510631 , China
| | - Yan-Qing Guan
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics , South China Normal University , Guangzhou 510631 , China
- Joint Laboratory of Laser Oncology with Cancer Center of Sun Yet-sen University , South China Normal University , Guangzhou 510631 , China
- School of Life Science , South China Normal University , Guangzhou 510631 , China
| |
Collapse
|