1
|
Zhao Y, Guo DF, Morgan DA, Cho YE, Rahmouni K. Adipocyte-specific disruption of the BBSome causes metabolic and autonomic dysfunction. Am J Physiol Regul Integr Comp Physiol 2024; 327:R54-R65. [PMID: 38738295 PMCID: PMC11380988 DOI: 10.1152/ajpregu.00039.2024] [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: 02/12/2024] [Revised: 04/16/2024] [Accepted: 05/03/2024] [Indexed: 05/14/2024]
Abstract
Obesity is a major public health issue due to its association with type 2 diabetes, hypertension, and other cardiovascular risks. The BBSome, a complex of eight conserved Bardet-Biedl syndrome (BBS) proteins, has emerged as a key regulator of energy and glucose homeostasis as well as cardiovascular function. However, the importance of adipocyte BBSome in controlling these physiological processes is not clear. Here, we show that adipocyte-specific constitutive disruption of the BBSome through selective deletion of the Bbs1 gene adiponectin (AdipoCre/Bbs1fl/fl mice) does not affect body weight under normal chow or high-fat and high-sucrose diet (HFHSD). However, constitutive BBSome deficiency caused impairment in glucose tolerance and insulin sensitivity. Similar phenotypes were observed after inducible adipocyte-specific disruption of the BBSome (AdipoCreERT2/Bbs1fl/fl mice). Interestingly, a significant increase in renal sympathetic nerve activity, measured using multifiber recording in the conscious state, was observed in AdipoCre/Bbs1fl/fl mice on both chow and HFHSD. A significant increase in tail-cuff arterial pressure was also observed in chow-fed AdipoCre/Bbs1fl/fl mice, but this was not reproduced when arterial pressure was measured by radiotelemetry. Moreover, AdipoCre/Bbs1fl/fl mice had no significant alterations in vascular reactivity. On the other hand, AdipoCre/Bbs1fl/fl mice displayed impaired baroreceptor reflex sensitivity when fed HFHSD, but not on normal chow. Taken together, these data highlight the relevance of the adipocyte BBSome for the regulation of glucose homeostasis and sympathetic traffic. The BBSome also contributes to baroreflex sensitivity under HFHSD, but not normal chow.NEW & NOTEWORTHY The current study show how genetic manipulation of fat cells impacts various functions of the body including sensitivity to the hormone insulin.
Collapse
Affiliation(s)
- Yuying Zhao
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States
- Interdisciplinary Graduate Program in Human Toxicology, Iowa City, Iowa, United States
| | - Deng-Fu Guo
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States
- Veterans Affairs Health Care System, Iowa City, Iowa, United States
| | - Donald A Morgan
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States
- Veterans Affairs Health Care System, Iowa City, Iowa, United States
| | - Young-Eun Cho
- College of Nursing, University of Iowa, Iowa City, Iowa, United States
| | - Kamal Rahmouni
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States
- Veterans Affairs Health Care System, Iowa City, Iowa, United States
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States
- Obesity Research and Education Initiative, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States
| |
Collapse
|
2
|
Türkkahraman D, Tekin S, Güllü M, Aykal G. Serum Ghrelin and Glucagon-like Peptide 1 Levels in Children with Prader-Willi and Bardet-Biedl Syndromes. J Clin Res Pediatr Endocrinol 2024; 16:146-150. [PMID: 38099591 DOI: 10.4274/jcrpe.galenos.2023.2023-7-7] [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] [Indexed: 06/04/2024] Open
Abstract
Objective Prader-Willi syndrome (PWS) and Bardet-Biedl syndrome (BBS) are causes of pediatric syndromic obesity. We aimed to investigate a possible role for ghrelin and glucagon-like peptide-1 (GLP-1) in the pathophysiology of PWS and BBS. Methods The study included 12 children with PWS, 12 children with BBS, 13 pediatric obese controls (OC) and 12 pediatric lean controls (LC). Fasting serum ghrelin and GLP-1 levels were measured by ELISA. Results In the PWS group, no significant difference was detected for median ghrelin levels when compared with OC and LC, which were 0.96 (0.69-1.15), 0.92 (0.72-1.20) and 1.13 (0.84-1.29) ng/mL, respectively. Similarly, there was no difference in PWS median GLP-1 levels when compared with OC and LC; 1.86 (1.5-2.94), 2.24 (1.62-2.78) and 2.06 (1.8-3.41) ng/mL, respectively. In the BBS group, there was no difference in median ghrelin levels when compared with OC and LC; 1.05 (0.87-1.51), 0.92 (0.72-1.20) and 1.13 (0.84-1.29) ng/mL, respectively. Neither was there a significant difference in median GLP-1 levels; 2.46 (1.91-4.17), 2.24 (1.62-2.78) and 2.06 (1.8-3.41) ng/mL for BBS, OC and LC, respectively. Conclusion There were no differences in median fasting ghrelin or GLP-1 levels when comparing patients with PWS and BBS with obese or lean peers. However, similar studies with larger series are needed.
Collapse
Affiliation(s)
- Doğa Türkkahraman
- University of Health Sciences Turkey, Antalya Training and Research Hospital, Clinic of Pediatric Endocrinology, Antalya, Turkey
| | - Suat Tekin
- University of Health Sciences Turkey, Antalya Training and Research Hospital, Clinic of Pediatric Endocrinology, Antalya, Turkey
| | - Merve Güllü
- University of Health Sciences Turkey, Antalya Training and Research Hospital, Clinic of Pediatric Endocrinology, Antalya, Turkey
| | - Güzin Aykal
- University of Health Sciences Turkey, Antalya Training and Research Hospital, Clinic of Biochemistry, Antalya, Turkey
| |
Collapse
|
3
|
Li MH, Chen IC, Yang HW, Yen HC, Huang YC, Hsu CC, Chen YM, Ke YY. The characterization and comorbidities of heterozygous Bardet-Biedl syndrome carriers. Int J Med Sci 2024; 21:784-794. [PMID: 38617006 PMCID: PMC11008491 DOI: 10.7150/ijms.92766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 02/10/2024] [Indexed: 04/16/2024] Open
Abstract
Introduction: Bardet-Biedl syndrome (BBS) is a rare autosomal recessive disorder with clinical features of retinal dystrophy, obesity, postaxial polydactyly, renal anomalies, learning disabilities, hypogonadism, and genitourinary abnormalities. Nevertheless, previous studies on the phenotypic traits of BBS heterozygous carriers have generated inconclusive results. The aim of our study was to investigate the impact of BBS heterozygosity on carriers when compared to non-carriers within the Taiwanese population. Materials and Methods: This study follows a hospital-based case-control design. We employed the Taiwan Biobank version 2 (TWBv2) array to identify three specific loci associated with BBS (rs773862084, rs567573386, and rs199910690). In total, 716 patients were included in the case group, and they were compared to a control group of 2,864 patients who lacked BBS alleles. The control group was selected through gender and age matching at a ratio of 1:4. The association between BBS-related loci and comorbidity was assessed using logistic regression models. Results: We found that BBS heterozygous carriers exhibited a significant association with elevated BMI levels, especially the variant rs199910690 in MKS1 (p=0.0037). The prevalence of comorbidities in the carriers' group was not higher than that in the non-carriers' group. Besides, the average values of the biochemistry data showed no significant differences, except for creatinine level. Furthermore, we conducted a BMI-based analysis to identify specific risk factors for chronic kidney disease (CKD). Our findings revealed that individuals carrying the CA/AA genotype of the BBS2 rs773862084 variant or the CT/TT genotype of the MKS1 rs199910690 variant showed a reduced risk of developing CKD, irrespective of their BMI levels. When stratified by BMI level, obese males with the MKS1 rs199910690 variant and obese females with the BBS2 rs773862084 variant exhibited a negative association with CKD development. Conclusion: We found that aside from the association with overweight and obesity, heterozygous BBS mutations did not appear to increase the predisposition of individuals to comorbidities and metabolic diseases. To gain a more comprehensive understanding of the genetic susceptibility associated with Bardet-Biedl Syndrome (BBS), further research is warranted.
Collapse
Affiliation(s)
- Meng-Hua Li
- Division of Pediatric Genetics and Metabolism, Children's Medical Center, Taichung Veterans General Hospital, Taichung, Taiwan
| | - I-Chieh Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Hui-Wen Yang
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Hsin-Chien Yen
- Division of Pediatric Genetics and Metabolism, Children's Medical Center, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yung-Chieh Huang
- Division of Nephrology, Department of Pediatrics, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chia-Chi Hsu
- Division of Pediatric Genetics and Metabolism, Children's Medical Center, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Ming Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taiwan
| | - Yu-Yuan Ke
- Division of Pediatric Genetics and Metabolism, Children's Medical Center, Taichung Veterans General Hospital, Taichung, Taiwan
| |
Collapse
|
4
|
Stump M, Guo DF, Rahmouni K. T cell-specific deficiency in BBSome component BBS1 interferes with selective immune responses. Am J Physiol Regul Integr Comp Physiol 2023; 324:R161-R170. [PMID: 36534590 PMCID: PMC9844976 DOI: 10.1152/ajpregu.00243.2022] [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: 09/28/2022] [Revised: 12/09/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022]
Abstract
Bsardet Biedl syndrome (BBS) is a genetic condition associated with various clinical features including cutaneous disorders and certain autoimmune and inflammatory diseases pointing to a potential role of BBS proteins in the regulation of immune function. BBS1 protein, which is a key component of the BBSome, a protein complex involved in the regulation of cilia function and other cellular processes, has been implicated in the immune synapse assembly by promoting the centrosome polarization to the antigen-presenting cells. Here, we assessed the effect of disrupting the BBSome, through Bbs1 gene deletion, in T cells. Interestingly, mice lacking the Bbs1 gene specifically in T cells (T-BBS1-/-) displayed normal body weight, adiposity, and glucose handling, but have smaller spleens. However, T-BBS1-/- mice had no change in the proportion and absolute number of B cells and T cells in the spleen and lymph nodes. There was also no alteration in the CD4/CD8 lineage commitment or survival in the thymus of T-BBS1-/- mice. On the other hand, T-BBS1-/- mice treated with Imiquimod dermally exhibited a significantly higher percentage of CD3-positive splenocytes that was due to CD4 but not CD8 T cell predominance. Notably, we found that T-BBS1-/- mice had significantly decreased wound closure, an effect that was more pronounced in males indicating that the BBSome plays an important role in T cell-mediated skin repair. Together, these findings implicate the BBSome in the regulation of selective functions of T cells.
Collapse
Affiliation(s)
- Madeliene Stump
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Physician Scientist Training Program, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Department of Dermatology, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Deng Fu Guo
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Veterans Affairs Health Care System, Iowa City, Iowa
| | - Kamal Rahmouni
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Veterans Affairs Health Care System, Iowa City, Iowa
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Obesity Research and Education Initiative, University of Iowa Carver College of Medicine, Iowa City, Iowa
| |
Collapse
|
5
|
Greeley SAW, Polak M, Njølstad PR, Barbetti F, Williams R, Castano L, Raile K, Chi DV, Habeb A, Hattersley AT, Codner E. ISPAD Clinical Practice Consensus Guidelines 2022: The diagnosis and management of monogenic diabetes in children and adolescents. Pediatr Diabetes 2022; 23:1188-1211. [PMID: 36537518 PMCID: PMC10107883 DOI: 10.1111/pedi.13426] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 12/24/2022] Open
Affiliation(s)
- Siri Atma W. Greeley
- Section of Pediatric and Adult Endocrinology, Diabetes and Metabolism, Kovler Diabetes Center and Comer Children's HospitalUniversity of Chicago MedicineChicagoIllinoisUSA
| | - Michel Polak
- Hôpital Universitaire Necker‐Enfants MaladesUniversité de Paris Cité, INSERM U1016, Institut IMAGINEParisFrance
| | - Pål R. Njølstad
- Department of Clinical ScienceUniversity of Bergen, and Children and Youth Clinic, Hauk eland University HospitalBergenNorway
| | - Fabrizio Barbetti
- Clinical Laboratory UnitBambino Gesù Children's Hospital, IRCCSRomeItaly
| | - Rachel Williams
- National Severe Insulin Resistance ServiceCambridge University Hospitals NHS TrustCambridgeUK
| | - Luis Castano
- Endocrinology and Diabetes Research Group, Biocruces Bizkaia Health Research InstituteCruces University Hospital, CIBERDEM, CIBERER, Endo‐ERN, UPV/EHUBarakaldoSpain
| | - Klemens Raile
- Department of Paediatric Endocrinology and DiabetologyCharité – UniversitätsmedizinBerlinGermany
| | - Dung Vu Chi
- Center for Endocrinology, Metabolism, Genetics and Molecular Therapy, Departement of Pediatric Endocrinology and DiabetesVietnam National Children's HospitalHanoiVietnam
- Department of Pediatrics and Department of Biology and Medical GeneticsHanoi Medical UniversityHanoiVietnam
| | - Abdelhadi Habeb
- Department of PediatricsPrince Mohamed bin Abdulaziz Hopsital, National Guard Health AffairsMadinahSaudi Arabia
| | - Andrew T. Hattersley
- Institute of Biomedical and Clinical SciencesUniversity of Exeter Medical SchoolExeterUK
| | - Ethel Codner
- Institute of Maternal and Child ResearchSchool of Medicine, University of ChileSantiagoChile
| |
Collapse
|
6
|
Loss of thyroid gland circadian PER2 rhythmicity in aged mice and its potential association with thyroid cancer development. Cell Death Dis 2022; 13:898. [PMID: 36284088 PMCID: PMC9596494 DOI: 10.1038/s41419-022-05342-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 10/04/2022] [Accepted: 10/12/2022] [Indexed: 11/28/2022]
Abstract
Molecular clocks operate in peripheral tissues, including endocrine glands, and play important regulatory roles in this context. However, potential age-related changes in the expression rhythmicity of clock genes and the effects of these changes on the thyroid gland remain unknown. In the present study, we evaluated the expression rhythmicity of peripheral thyroid clock genes in aged mice using RNA-seq transcriptomic analysis in young (3.5-month) versus aged (20-month) mice. In addition, we determined the cellular effects of silencing of PER2, a major clock gene regulator, in human thyroid cell lines. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis revealed that differentially expressed genes (DEGs) in the thyroid glands of aged mice were involved in mitogen-activated protein kinase (MAPK) signaling, chemokine signaling, circadian entrainment, PI3K/AKT signaling, and Apelin signaling. The expression of circadian clock genes Arntl/Bmal1 was significantly downregulated in thyroid glands of aged mice, whereas the expression of genes involved in regulation of cell proliferation, migration, and tumorigenesis was upregulated. Peripheral thyroid clock genes, particularly Per mRNA and PER2 protein, were downregulated in the thyroid glands of aged mice, and circadian oscillation of these genes was declined. Knockdown of the circadian clock gene PER2 in human thyroid follicular cells induced AP-1 activity via JNK MAPK signaling activation, which increased cell proliferation. Furthermore, the aging-related loss of PER2 circadian oscillation activated the AP-1 transcription factor via the JNK MAPK pathway, which could contribute to thyroid hyperplasia, a common age-related condition.
Collapse
|
7
|
Shalitin S, Gat-Yablonski G. Associations of Obesity with Linear Growth and Puberty. Horm Res Paediatr 2022; 95:120-136. [PMID: 34130293 DOI: 10.1159/000516171] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 03/27/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The prevalence of obesity in childhood has increased dramatically in recent decades with increased risk of developing cardiometabolic and other comorbidities. Childhood adiposity may also influence processes of growth and puberty. SUMMARY Growth patterns of obesity during childhood have been shown to be associated with increased linear growth in early childhood, leading to accelerated epiphyseal growth plate (EGP) maturation. Several hormones secreted by the adipose tissue may affect linear growth in the context of obesity, both via the growth hormone IGF-1 axis and via a direct effect on the EGP. The observation that children with obesity tend to mature earlier than lean children has led to the assumption that the degree of body fatness may trigger the neuroendocrine events that lead to pubertal onset. The most probable link between obesity and puberty is leptin and its interaction with the kisspeptin system, which is an important regulator of puberty. However, peripheral action of adipose tissue could also be involved in changes in the onset of puberty. In addition, nutritional factors, epigenetics, and endocrine-disrupting chemicals are potential mediators linking pubertal onset to obesity. In this review, we focused on interactions of obesity with linear growth and pubertal processes, based on basic research and clinical data in humans. KEY MESSAGE Children with obesity are subject to accelerated linear growth with risk of impaired adult height and early puberty, with its psychological consequences. The data highlight another important objective in combatting childhood obesity, for the prevention of abnormal growth and pubertal patterns.
Collapse
Affiliation(s)
- Shlomit Shalitin
- National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, The Jesse Z and Sara Lea Shafer Institute for Endocrinology and Diabetes, Petach Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Galia Gat-Yablonski
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Felsenstein Medical Research Center, Petach Tikva, Israel
| |
Collapse
|
8
|
Roberts KJ, Ariza AJ, Selvaraj K, Quadri M, Mangarelli C, Neault S, Davis EE, Binns HJ. Testing for rare genetic causes of obesity: findings and experiences from a pediatric weight management program. Int J Obes (Lond) 2022; 46:1493-1501. [PMID: 35562395 PMCID: PMC9105591 DOI: 10.1038/s41366-022-01139-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Genetic screening for youth with obesity in the absence of syndromic findings has not been part of obesity management. For children with early onset obesity, genetic screening is recommended for those having clinical features of genetic obesity syndromes (including hyperphagia). OBJECTIVES The overarching goal of this work is to report the findings and experiences from one pediatric weight management program that implemented targeted sequencing analysis for genes known to cause rare genetic disorders of obesity. SUBJECTS/METHODS This exploratory study evaluated youth tested over an 18-month period using a panel of 40-genes in the melanocortin 4 receptor pathway. Medical records were reviewed for demographic and visit information, including body mass index (BMI) percent of 95th percentile (%BMIp95) and two eating behaviors. RESULTS Of 117 subjects: 51.3% were male; 53.8% Hispanic; mean age 10.2 years (SD 3.8); mean %BMIp95 157% (SD 29%). Most subjects were self- or caregiver-reported to have overeating to excess or binge eating (80.3%) and sneaking food or eating in secret (59.0%). Among analyzed genes, 72 subjects (61.5%) had at least one variant reported; 50 (42.7%) had a single variant reported; 22 (18.8%) had 2-4 variants reported; most variants were rare (<0.05% minor allele frequency [MAF]), and of uncertain significance; all variants were heterozygous. Nine subjects (7.7%) had a variant reported as PSCK1 "risk" or MC4R "likely pathogenic"; 39 (33.3%) had a Bardet-Biedl Syndrome (BBS) gene variant (4 with "pathogenic" or "likely pathogenic" variants). Therefore, 9 youth (7.7%) had gene variants previously identified as increasing risk for obesity and 4 youth (3.4%) had BBS carrier status. CONCLUSIONS Panel testing identified rare variants of uncertain significance in most youth tested, and infrequently identified variants previously reported to increase the risk for obesity. Further research in larger cohorts is needed to understand how genetic variants influence the expression of non-syndromic obesity.
Collapse
Affiliation(s)
- Karyn J Roberts
- College of Nursing, University of Wisconsin-Milwaukee, PO Box 413, Milwaukee, WI, 53201-0413, USA. .,Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.
| | - Adolfo J Ariza
- Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.,Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA.,Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Kavitha Selvaraj
- Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.,Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Maheen Quadri
- Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.,Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Caren Mangarelli
- Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.,Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Sarah Neault
- Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Erica E Davis
- Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.,Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Helen J Binns
- Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.,Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA.,Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| |
Collapse
|
9
|
Bardet-Biedl Syndrome-Multiple Kaleidoscope Images: Insight into Mechanisms of Genotype-Phenotype Correlations. Genes (Basel) 2021; 12:genes12091353. [PMID: 34573333 PMCID: PMC8465569 DOI: 10.3390/genes12091353] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/22/2021] [Accepted: 08/27/2021] [Indexed: 01/04/2023] Open
Abstract
Bardet-Biedl Syndrome is a rare non-motile primary ciliopathy with multisystem involvement and autosomal recessive inheritance. The clinical picture is extremely polymorphic. The main clinical features are retinal cone-rod dystrophy, central obesity, postaxial polydactyly, cognitive impairment, hypogonadism and genitourinary abnormalities, and kidney disease. It is caused by various types of mutations, mainly in genes encoding BBSome proteins, chaperonins, and IFT complex. Variable expressivity and pleiotropy are correlated with the existence of multiple genes and variants modifiers. This review is focused on the phenomena of heterogeneity (locus, allelic, mutational, and clinical) in Bardet-Biedl Syndrome, its mechanisms, and importance in early diagnosis and proper management.
Collapse
|
10
|
Islam S, Reddy UK, Natarajan P, Abburi VL, Bajwa AA, Imran M, Zahoor MY, Abdullah M, Bukhari AM, Iqbal S, Ashraf K, Nadeem A, Rehman H, Rashid I, Shehzad W. Population demographic history and population structure for Pakistani Nili-Ravi breeding bulls based on SNP genotyping to identify genomic regions associated with male effects for milk yield and body weight. PLoS One 2020; 15:e0242500. [PMID: 33232358 PMCID: PMC7685427 DOI: 10.1371/journal.pone.0242500] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 11/03/2020] [Indexed: 11/20/2022] Open
Abstract
The domestic Nili-Ravi water buffalo (Bubalus bubalis) is the best dairy animal contributing 68% to total milk production in Pakistan. In this study, we identified genome-wide single nucleotide polymorphisms (SNPs) to estimate various population genetic parameters such as diversity, pairwise population differentiation, linkage disequilibrium (LD) distribution and for genome-wide association study for milk yield and body weight traits in the Nili-Ravi dairy bulls that they may pass on to their daughters who are retained for milking purposes. The genotyping by sequencing approach revealed 13,039 reference genome-anchored SNPs with minor allele frequency of 0.05 among 167 buffalos. Population structure analysis revealed that the bulls were grouped into two clusters (K = 2), which indicates the presence of two different lineages in the Pakistani Nili-Ravi water buffalo population, and we showed the extent of admixture of these two lineages in our bull collection. LD analysis revealed 4169 significant SNP associations, with an average LD decay of 90 kb for these buffalo genome. Genome-wide association study involved a multi-locus mixed linear model for milk yield and body weight to identify genome-wide male effects. Our study further illustrates the utility of the genotyping by sequencing approach for identifying genomic regions to uncover additional demographic complexity and to improve the complex dairy traits of the Pakistani Nili-Ravi water buffalo population that would provide the lot of economic benefits to dairy industry.
Collapse
Affiliation(s)
- Saher Islam
- Institute of Biochemistry and Biotechnology, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Umesh K. Reddy
- Department of Biology, West Virginia State University, Institute, West Virginia, United States of America
| | - Purushothaman Natarajan
- Department of Biology, West Virginia State University, Institute, West Virginia, United States of America
| | - Venkata Lakshmi Abburi
- Department of Biology, West Virginia State University, Institute, West Virginia, United States of America
| | - Amna Arshad Bajwa
- Institute of Biochemistry and Biotechnology, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Muhammad Imran
- Institute of Biochemistry and Biotechnology, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Muhammad Yasir Zahoor
- Institute of Biochemistry and Biotechnology, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Muhammad Abdullah
- Department of Livestock Production, University of Veterinary and Animal Sciences, Pattoki, Pakistan
| | - Aamir Mehmood Bukhari
- Semen Production Unit, Qadirabad, District Sahiwal, Pakistan
- Livestock and Dairy Development Department, Government of the Punjab, Lahore, Pakistan
| | - Sajid Iqbal
- Semen Production Unit, Qadirabad, District Sahiwal, Pakistan
- Livestock and Dairy Development Department, Government of the Punjab, Lahore, Pakistan
| | - Kamran Ashraf
- Department of Parasitology, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Asif Nadeem
- Institute of Biochemistry and Biotechnology, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Habibur Rehman
- Department of Physiology, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Imran Rashid
- Department of Parasitology, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Wasim Shehzad
- Institute of Biochemistry and Biotechnology, University of Veterinary and Animal Sciences, Lahore, Pakistan
| |
Collapse
|
11
|
Haws R, Brady S, Davis E, Fletty K, Yuan G, Gordon G, Stewart M, Yanovski J. Effect of setmelanotide, a melanocortin-4 receptor agonist, on obesity in Bardet-Biedl syndrome. Diabetes Obes Metab 2020; 22:2133-2140. [PMID: 32627316 PMCID: PMC7689750 DOI: 10.1111/dom.14133] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/25/2020] [Accepted: 06/30/2020] [Indexed: 12/16/2022]
Abstract
AIM To report an analysis of ~1 year of setmelanotide treatment for obesity and hunger, as well as metabolic and cardiac outcomes, in individuals with Bardet-Biedl syndrome (BBS). MATERIALS AND METHODS Individuals aged 12 years and older with BBS received once-daily setmelanotide. The dose was titrated every 2 weeks to establish the individual therapeutic dose (≤3 mg); treatment continued for an additional 10 weeks. Participants who lost 5 kg or more (or ≥5% of body weight if <100 kg at baseline) continued into the 52-week extension phase. The primary outcome was mean percent change from baseline in body weight at 3 months. Hunger scores and safety were secondary outcomes. RESULTS From February 2017 and February 2018, 10 individuals were screened; eight completed the 3-month treatment phase and seven completed the extension phase. Mean percent change in body weight from baseline to 3 months was -5.5% (90% CI, -9.3% to -1.6%; n = 8); change from baseline was -11.3% (90% CI, -15.5% to -7.0%; n = 8) at 6 months and -16.3% (90% CI, -19.9% to -12.8%; n = 7) at 12 months. All participants reported at least one treatment-emergent adverse event (AE), most commonly injection-site reaction. No AEs led to study withdrawal or death. Most, morning, and average hunger scores were reduced across time points. CONCLUSIONS Setmelanotide reduced body weight and hunger in individuals with BBS and had a safety profile consistent with previous reports. Setmelanotide may be a treatment option in individuals with BBS-associated obesity and hyperphagia.
Collapse
Affiliation(s)
- Robert Haws
- Marshfield Clinic Research InstituteMarshfieldWisconsinUSA
| | - Sheila Brady
- Section on Growth and ObesityEunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaMarylandUSA
| | - Elisabeth Davis
- Section on Growth and ObesityEunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaMarylandUSA
| | | | - Guojun Yuan
- Rhythm PharmaceuticalsBostonMassachusettsUSA
| | | | | | - Jack Yanovski
- Section on Growth and ObesityEunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
12
|
Suciu SK, Caspary T. Cilia, neural development and disease. Semin Cell Dev Biol 2020; 110:34-42. [PMID: 32732132 DOI: 10.1016/j.semcdb.2020.07.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/17/2020] [Accepted: 07/18/2020] [Indexed: 12/16/2022]
Abstract
Neural development requires a series of cellular events starting with cell specification, proliferation, and migration. Subsequently, axons and dendrites project from the cell surface to form connections to other neurons, interneurons and glia. Anomalies in any one of these steps can lead to malformation or malfunction of the nervous system. Here we review the critical role the primary cilium plays in the fundamental steps of neurodevelopment. By highlighting human diseases caused by mutations in cilia-associated proteins, it is clear that cilia are essential to multiple neural processes. Furthermore, we explore whether additional aspects of cilia regulation, most notably post-translational modification of the tubulin scaffold in cilia, play underappreciated roles in neural development. Finally, we discuss whether cilia-associated proteins function outside the cilium in some aspects of neurodevelopment. These data underscore both the importance of cilia in the nervous system and some outstanding questions in the field.
Collapse
Affiliation(s)
- Sarah K Suciu
- Genetics and Molecular Biology Graduate Program, USA; Department of Human Genetics, Emory University, Atlanta, GA 30322, Georgia
| | - Tamara Caspary
- Department of Human Genetics, Emory University, Atlanta, GA 30322, Georgia.
| |
Collapse
|
13
|
Ruiz-Gayo M, Olmo ND. Interaction Between Circadian Rhythms, Energy Metabolism, and Cognitive Function. Curr Pharm Des 2020; 26:2416-2425. [PMID: 32156228 DOI: 10.2174/1381612826666200310145006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 02/11/2020] [Indexed: 11/22/2022]
Abstract
The interaction between meal timing and light regulates circadian rhythms in mammals and not only determines the sleep-wake pattern but also the activity of the endocrine system. Related with that, the necessity to fulfill energy needs is a driving force that requires the participation of cognitive skills whose performance has been shown to undergo circadian variations. These facts have led to the concept that cognition and feeding behaviour can be analysed from a chronobiological perspective. In this context, research carried out during the last two decades has evidenced the link between feeding behaviour/nutritional habits and cognitive processes, and has highlighted the impact of circadian disorders on cognitive decline. All that has allowed hypothesizing a tight relationship between nutritional factors, chronobiology, and cognition. In this connection, experimental diets containing elevated amounts of fat and sugar (high-fat diets; HFDs) have been shown to alter in rodents the circadian distribution of meals, and to have a negative impact on cognition and motivational aspects of behaviour that disappear when animals are forced to adhere to a standard temporal eating pattern. In this review, we will present relevant studies focussing on the effect of HFDs on cognitive aspects of behaviour, paying particular attention to the influence that chronobiological alterations caused by these diets may have on hippocampaldependent cognition.
Collapse
Affiliation(s)
- Mariano Ruiz-Gayo
- Department of Health and Pharmaceutical Sciences, School of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Nuria D Olmo
- Department of Health and Pharmaceutical Sciences, School of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| |
Collapse
|
14
|
Abstract
The BBSome, a complex of 8 BBS (Bardet-Biedl syndrome) proteins known for its role in the control of cilia function and other cellular processes, has been implicated in blood pressure control, but the underlying mechanisms are not fully understood. Here, we show that neuronal BBSome plays an important role in blood pressure regulation. Targeted inactivation of the BBSome in the nervous system through Bbs1 gene deletion causes sympathetically mediated increase in blood pressure in mice. This phenotype is reproduced by selective ablation of the Bbs1 gene from the LRb (leptin receptor)-expressing neurons. Strikingly, the well-known role of the BBSome in the regulation of cilia formation and function is unlikely to account for the prohypertensive effect of BBSome inactivation as disruption of the IFT (intraflagellar transport) machinery required for ciliogenesis by deleting the Ift88 gene in LRb neurons had no effect on arterial pressure and sympathetic nerve activity. Furthermore, we found that Bbs1 gene deletion from AgRP (agouti-related protein) neurons or POMC (proopiomelanocortin) neurons increased renal and splanchnic sympathetic nerve activity without altering blood pressure. This lack of blood pressure increase despite the sympathetic overdrive may be explained by vascular adrenergic desensitization as indicated by the reduced vascular contractile response evoked by phenylephrine and the decreased expression of adrenergic receptors. Our results identify the neuronal BBSome as a new player in hemodynamic, sympathetic, and vascular regulation, in a manner independent of cilia.
Collapse
Affiliation(s)
- Deng-Fu Guo
- From the Department of Neuroscience and Pharmacology (D.-F.G., J.J.R., D.A.M., K.R.), University of Iowa Carver College of Medicine, Iowa City
| | - John J Reho
- From the Department of Neuroscience and Pharmacology (D.-F.G., J.J.R., D.A.M., K.R.), University of Iowa Carver College of Medicine, Iowa City
| | - Donald A Morgan
- From the Department of Neuroscience and Pharmacology (D.-F.G., J.J.R., D.A.M., K.R.), University of Iowa Carver College of Medicine, Iowa City
| | - Kamal Rahmouni
- From the Department of Neuroscience and Pharmacology (D.-F.G., J.J.R., D.A.M., K.R.), University of Iowa Carver College of Medicine, Iowa City.,Department of Internal Medicine (K.R.), University of Iowa Carver College of Medicine, Iowa City.,Obesity Research and Education Initiative (K.R.), University of Iowa Carver College of Medicine, Iowa City.,Fraternal Order of Eagles Diabetes Research Center (K.R.), University of Iowa Carver College of Medicine, Iowa City.,Iowa Neuroscience Institute (K.R.), University of Iowa Carver College of Medicine, Iowa City.,Veterans Affairs Health Care System, Iowa City (K.R.)
| |
Collapse
|
15
|
Pottorf TS, Fagan MP, Burkey BF, Cho DJ, Vath JE, Tran PV. MetAP2 inhibition reduces food intake and body weight in a ciliopathy mouse model of obesity. JCI Insight 2020; 5:134278. [PMID: 31877115 DOI: 10.1172/jci.insight.134278] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/18/2019] [Indexed: 12/13/2022] Open
Abstract
The ciliopathies Bardet-Biedl syndrome and Alström syndrome are genetically inherited pleiotropic disorders with hyperphagia and obesity as primary clinical features. Methionine aminopeptidase 2 inhibitors (MetAP2i) have been shown in preclinical and clinical studies to reduce food intake, body weight, and adiposity. Here, we investigated the effects of MetAP2i administration in a mouse model of ciliopathy produced by conditional deletion of the Thm1 gene in adulthood. Thm1 conditional knockout (cko) mice showed decreased hypothalamic proopiomelanocortin expression as well as hyperphagia, obesity, metabolic disease, and hepatic steatosis. In obese Thm1-cko mice, 2-week administration of MetAP2i reduced daily food intake and reduced body weight 17.1% from baseline (vs. 5% reduction for vehicle). This was accompanied by decreased levels of blood glucose, insulin, and leptin. Further, MetAP2i reduced gonadal adipose depots and adipocyte size and improved liver morphology. This is the first report to our knowledge of MetAP2i reducing hyperphagia and body weight and ameliorating metabolic indices in a mouse model of ciliopathy. These results support further investigation of MetAP2 inhibition as a potential therapeutic strategy for ciliary-mediated forms of obesity.
Collapse
Affiliation(s)
- Tana S Pottorf
- Jared Grantham Kidney Institute and.,Department of Anatomy and Cell Biology, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
| | | | | | - David J Cho
- Jared Grantham Kidney Institute and.,Department of Anatomy and Cell Biology, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
| | | | - Pamela V Tran
- Jared Grantham Kidney Institute and.,Department of Anatomy and Cell Biology, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
| |
Collapse
|
16
|
Singh SK, Gui M, Koh F, Yip MC, Brown A. Structure and activation mechanism of the BBSome membrane protein trafficking complex. eLife 2020; 9:53322. [PMID: 31939736 PMCID: PMC7018513 DOI: 10.7554/elife.53322] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 01/06/2020] [Indexed: 12/19/2022] Open
Abstract
Bardet-Biedl syndrome (BBS) is a currently incurable ciliopathy caused by the failure to correctly establish or maintain cilia-dependent signaling pathways. Eight proteins associated with BBS assemble into the BBSome, a key regulator of the ciliary membrane proteome. We report the electron cryomicroscopy (cryo-EM) structures of the native bovine BBSome in inactive and active states at 3.1 and 3.5 Å resolution, respectively. In the active state, the BBSome is bound to an Arf-family GTPase (ARL6/BBS3) that recruits the BBSome to ciliary membranes. ARL6 recognizes a composite binding site formed by BBS1 and BBS7 that is occluded in the inactive state. Activation requires an unexpected swiveling of the β-propeller domain of BBS1, the subunit most frequently implicated in substrate recognition, which widens a central cavity of the BBSome. Structural mapping of disease-causing mutations suggests that pathogenesis results from folding defects and the disruption of autoinhibition and activation.
Collapse
Affiliation(s)
- Sandeep K Singh
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, United States
| | - Miao Gui
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, United States
| | - Fujiet Koh
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, United States
| | - Matthew Cj Yip
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, United States
| | - Alan Brown
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, United States
| |
Collapse
|
17
|
Horiuchi K, Kogiso T, Sagawa T, Ito T, Taniai M, Miura K, Hattori M, Morisada N, Hashimoto E, Tokushige K. Bardet-Biedl Syndrome Caused by Skipping of SCLT1 Complicated by Microvesicular Steatohepatitis. Intern Med 2020; 59:2719-2724. [PMID: 33132306 PMCID: PMC7691027 DOI: 10.2169/internalmedicine.5045-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
We treated the case of a 22-year-old male patient with liver dysfunction. At 1 year of age, hepatic fibrosis was suspected. In addition, due to the presence of retinitis pigmentosa, renal failure, obesity, mental retardation, and hypogonadism, he was diagnosed with Bardet-Biedl syndrome (BBS). Skipping of exons 14 and 17 in the sodium channel and clathrin linker 1 (SCLT1) gene was observed. At 22 years of age, the liver enzyme levels were further elevated and a diagnosis of microvesicular steatohepatitis was made. Insulin resistance, a reduction of muscle mass, an impairment of the fatty acid metabolism, and hyperleptinemia in this syndrome may cause steatohepatitis.
Collapse
Affiliation(s)
- Kentaro Horiuchi
- Institute of Gastroenterology, Department of Internal Medicine, Tokyo Women's Medical University, Japan
| | - Tomomi Kogiso
- Institute of Gastroenterology, Department of Internal Medicine, Tokyo Women's Medical University, Japan
| | - Takaomi Sagawa
- Institute of Gastroenterology, Department of Internal Medicine, Tokyo Women's Medical University, Japan
| | - Taito Ito
- Institute of Gastroenterology, Department of Internal Medicine, Tokyo Women's Medical University, Japan
| | - Makiko Taniai
- Institute of Gastroenterology, Department of Internal Medicine, Tokyo Women's Medical University, Japan
| | - Kenichiro Miura
- Department of Pediatric Nephrology, Tokyo Women's Medical University, Japan
| | - Motoshi Hattori
- Department of Pediatric Nephrology, Tokyo Women's Medical University, Japan
| | - Naoya Morisada
- Department of Pediatrics, Kobe University Graduate School of Medicine, Japan
| | | | - Katsutoshi Tokushige
- Institute of Gastroenterology, Department of Internal Medicine, Tokyo Women's Medical University, Japan
| |
Collapse
|
18
|
Jacobs DT, Allard BA, Pottorf TS, Silva LM, Wang W, Al-Naamani A, Agborbesong E, Wang T, Carr DA, Tran PV. Intraflagellar-transport A dysfunction causes hyperphagia-induced systemic insulin resistance in a pre-obese state. FASEB J 2019; 34:148-160. [PMID: 31914634 DOI: 10.1096/fj.201900751r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 10/04/2019] [Accepted: 10/21/2019] [Indexed: 12/13/2022]
Abstract
Deletion of murine Thm1, an intraflagellar transport A (IFT-A) component that mediates ciliary protein trafficking, causes hyperphagia, obesity, and metabolic syndrome. The role of Thm1 or IFT-A in adipogenesis and insulin sensitivity is unknown. Here, we report that Thm1 knockdown in 3T3-L1 pre-adipocytes promotes adipogenesis and enhances insulin sensitivity in vitro. Yet, pre-obese Thm1 conditional knockout mice show systemic insulin resistance. While insulin-induced AKT activation in Thm1 mutant adipose depots and skeletal muscle are similar to those of control littermates, an attenuated insulin response arises in the mutant liver. Insulin treatment of control and Thm1 mutant primary hepatocytes results in similar AKT activation. Moreover, pair-feeding Thm1 conditional knockout mice produces a normal insulin response, both in the liver and systemically. Thus, hyperphagia caused by a cilia defect, induces hepatic insulin resistance via a non-cell autonomous mechanism. In turn, hepatic insulin resistance drives systemic insulin resistance prior to an obese phenotype. These data demonstrate that insulin signaling across cell types is regulated differentially, and that the liver is particularly susceptible to hyperphagia-induced insulin resistance and a critical determinant of systemic insulin resistance.
Collapse
Affiliation(s)
- Damon T Jacobs
- Department of Anatomy and Cell Biology, Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Bailey A Allard
- Department of Anatomy and Cell Biology, Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Tana S Pottorf
- Department of Anatomy and Cell Biology, Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Luciane M Silva
- Department of Anatomy and Cell Biology, Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Wei Wang
- Department of Anatomy and Cell Biology, Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Aisha Al-Naamani
- Department of Anatomy and Cell Biology, Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Ewud Agborbesong
- Department of Anatomy and Cell Biology, Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Tao Wang
- Department of Anatomy and Cell Biology, Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Dajanae A Carr
- Department of Anatomy and Cell Biology, Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Pamela V Tran
- Department of Anatomy and Cell Biology, Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
19
|
Reho JJ, Guo DF, Morgan DA, Rahmouni K. Smooth Muscle Cell-Specific Disruption of the BBSome Causes Vascular Dysfunction. Hypertension 2019; 74:817-825. [PMID: 31422694 DOI: 10.1161/hypertensionaha.119.13382] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The BBSome-a complex consisting of 8 Bardet-Biedl syndrome proteins-is involved in the regulation of various cellular processes. Recently, the BBSome complex has emerged as an important regulator of cardiovascular function with implications for disease. In this study, we examined the role of the BBSome in vascular smooth muscle and its effects on the regulation of cardiovascular function. Smooth muscle-specific disruption of the BBSome through tamoxifen-inducible deletion of Bbs1 gene-a critical component of the BBSome complex-reduces relaxation and enhances contractility of vascular rings and increases aortic stiffness independent of changes in arterial blood pressure. Mechanistically, we demonstrate that smooth muscle Bbs1 gene deletion increases vascular angiotensinogen gene expression implicating the renin-angiotensin system in these altered cardiovascular responses. Additionally, we report that smooth muscle-specific Bbs1 knockout mice demonstrate enhanced ET-1 (endothelin-1)-induced contractility of mesenteric arteries-an effect reversed by blockade of the AT1 (angiotensin type 1 receptor) with losartan. These findings highlight the importance of the smooth muscle BBSome in the control of vascular function and arterial stiffness through modulation of renin-angiotensin system signaling.
Collapse
Affiliation(s)
- John J Reho
- From the Department of Pharmacology (J.J.R., D.-F.G., D.A.M., K.R.), University of Iowa Carver College of Medicine
| | - Deng-Fu Guo
- From the Department of Pharmacology (J.J.R., D.-F.G., D.A.M., K.R.), University of Iowa Carver College of Medicine
| | - Donald A Morgan
- From the Department of Pharmacology (J.J.R., D.-F.G., D.A.M., K.R.), University of Iowa Carver College of Medicine
| | - Kamal Rahmouni
- From the Department of Pharmacology (J.J.R., D.-F.G., D.A.M., K.R.), University of Iowa Carver College of Medicine.,Department of Internal Medicine (K.R.), University of Iowa Carver College of Medicine.,Obesity Education and Research Initiative (K.R.), University of Iowa Carver College of Medicine.,Fraternal Order of Eagles Diabetes Research Center (K.R.), University of Iowa Carver College of Medicine.,Veterans Affairs Health Care System, Iowa City, IA (K.R.)
| |
Collapse
|
20
|
Guo DF, Lin Z, Wu Y, Searby C, Thedens DR, Richerson GB, Usachev YM, Grobe JL, Sheffield VC, Rahmouni K. The BBSome in POMC and AgRP Neurons Is Necessary for Body Weight Regulation and Sorting of Metabolic Receptors. Diabetes 2019; 68:1591-1603. [PMID: 31127052 PMCID: PMC6692817 DOI: 10.2337/db18-1088] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 05/12/2019] [Indexed: 12/22/2022]
Abstract
The BBSome, a complex of eight Bardet-Biedl syndrome (BBS) proteins involved in cilia function, has emerged as an important regulator of energy balance, but the underlying cellular and molecular mechanisms are not fully understood. Here, we show that the control of energy homeostasis by the anorexigenic proopiomelanocortin (POMC) neurons and orexigenic agouti-related peptide (AgRP) neurons require intact BBSome. Targeted disruption of the BBSome by Bbs1 gene deletion in POMC or AgRP neurons increases body weight and adiposity. We demonstrate that obesity in mice lacking the Bbs1 gene in POMC neurons is associated with hyperphagia. Mechanistically, we present evidence implicating the BBSome in the trafficking of G protein-coupled neuropeptide Y Y2 receptor (NPY2R) and serotonin 5-hydroxytryptamine (HT)2C receptor (5-HT2CR) to cilia and plasma membrane, respectively. Consistent with this, loss of the BBSome reduced cell surface expression of the 5-HT2CR, interfered with serotonin-evoked increase in intracellular calcium and membrane potential, and blunted the anorectic and weight-reducing responses evoked by the 5-HT2cR agonist, lorcaserin. Finally, we show that disruption of the BBSome causes the 5-HT2CR to be stalled in the late endosome. Our results demonstrate the significance of the hypothalamic BBSome for the control of energy balance through regulation of trafficking of important metabolic receptors.
Collapse
Affiliation(s)
- Deng-Fu Guo
- Department of Pharmacology, University of Iowa, Iowa City, IA
| | - Zhihong Lin
- Department of Pharmacology, University of Iowa, Iowa City, IA
| | - Yuanming Wu
- Department of Neurology, University of Iowa, Iowa City, IA
| | - Charles Searby
- Department of Pediatrics, University of Iowa, Iowa City, IA
| | | | | | - Yuriy M Usachev
- Department of Pharmacology, University of Iowa, Iowa City, IA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA
| | - Justin L Grobe
- Department of Pharmacology, University of Iowa, Iowa City, IA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA
- Obesity Education and Research Initiative, University of Iowa, Iowa City, IA
| | - Val C Sheffield
- Department of Pediatrics, University of Iowa, Iowa City, IA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA
- Obesity Education and Research Initiative, University of Iowa, Iowa City, IA
| | - Kamal Rahmouni
- Department of Pharmacology, University of Iowa, Iowa City, IA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA
- Obesity Education and Research Initiative, University of Iowa, Iowa City, IA
- Department of Internal Medicine, University of Iowa, Iowa City, IA
| |
Collapse
|
21
|
Del Olmo N, Ruiz-Gayo M. Influence of High-Fat Diets Consumed During the Juvenile Period on Hippocampal Morphology and Function. Front Cell Neurosci 2018; 12:439. [PMID: 30515083 PMCID: PMC6255817 DOI: 10.3389/fncel.2018.00439] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 11/05/2018] [Indexed: 12/18/2022] Open
Abstract
The negative impact of obesity on neurocognitive functioning is an issue of increasing clinical interest. Over the last decade, a number of studies have analyzed the influence of high-fat diets (HFDs) on cognitive performance, particularly in adolescent individuals. Different approaches, including behavioral, neurochemical, electrophysiological and morphological studies, have been developed to address the effect of HFDs on neural processes interfering with learning and memory skills in rodents. Many of the studies have focused on learning and memory related to the hippocampus and the mechanisms underlying these processes. The goal of the current review article is to highlight the relationship between hippocampal learning/memory deficits and nutritional/endocrine inputs derived from HFDs consumption, with a special emphasis on research showing the effect of HFDs intake during the juvenile period. We have also reviewed recent research regarding the effect of HFDs on hippocampal neurotransmission. An overview of research suggesting the involvement of fatty acid (FA) receptor-mediated signaling pathways in memory deficits triggered by HFDs is also provided. Finally, the role of leptin and HFD-evoked hyperleptinemia is discussed.
Collapse
Affiliation(s)
- Nuria Del Olmo
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
| | - Mariano Ruiz-Gayo
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
| |
Collapse
|
22
|
Lewis WR, Bales KL, Revell DZ, Croyle MJ, Engle SE, Song CJ, Malarkey EB, Uytingco CR, Shan D, Antonellis PJ, Nagy TR, Kesterson RA, Mrug MM, Martens JR, Berbari NF, Gross AK, Yoder BK. Mks6 mutations reveal tissue- and cell type-specific roles for the cilia transition zone. FASEB J 2018; 33:1440-1455. [PMID: 30133325 PMCID: PMC6355093 DOI: 10.1096/fj.201801149r] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The transition zone (TZ) is a domain at the base of the cilium that is involved in maintaining ciliary compartment-specific sensory and signaling activity by regulating cilia protein composition. Mutations in TZ proteins result in cilia dysfunction, often causing pleiotropic effects observed in a group of human diseases classified as ciliopathies. The purpose of this study is to describe the importance of the TZ component Meckel-Grüber syndrome 6 ( Mks6) in several organ systems and tissues regarding ciliogenesis and cilia maintenance using congenital and conditional mutant mouse models. Similar to MKS, congenital loss of Mks6 is embryonic lethal, displaying cilia loss and altered cytoskeletal microtubule modifications but only in specific cell types. Conditional Mks6 mutants have a variable cystic kidney phenotype along with severe retinal degeneration with mislocalization of phototransduction cascade proteins. However, other phenotypes, such as anosmia and obesity, which are typically associated with cilia and TZ dysfunction, were not evident. These data indicate that despite Mks6 being a core TZ component, it has tissue- or cell type-specific functions important for cilia formation and cilia sensory and signaling activities. Lewis, W. R., Bales, K. L., Revell, D. Z., Croyle, M. J., Engle, S. E., Song, C. J., Malarkey, E. B., Uytingco, C. R., Shan, D., Antonellis, P. J., Nagy, T. R., Kesterson, R. A., Mrug, M. M., Martens, J. R., Berbari, N. F., Gross, A. K., Yoder, B. K. Mks6 mutations reveal tissue- and cell type-specific roles for the cilia transition zone.
Collapse
Affiliation(s)
- Wesley R Lewis
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Katie L Bales
- Department of Optometry and Vision Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Dustin Z Revell
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Mandy J Croyle
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Staci E Engle
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, USA
| | - Cheng Jack Song
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Erik B Malarkey
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Cedric R Uytingco
- Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Dan Shan
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Patrick J Antonellis
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, USA
| | - Tim R Nagy
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA; and
| | - Robert A Kesterson
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Michal M Mrug
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jeffrey R Martens
- Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Nicolas F Berbari
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, USA
| | - Alecia K Gross
- Department of Optometry and Vision Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Bradley K Yoder
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
23
|
Weihbrecht K, Goar WA, Pak T, Garrison JE, DeLuca AP, Stone EM, Scheetz TE, Sheffield VC. Keeping an Eye on Bardet-Biedl Syndrome: A Comprehensive Review of the Role of Bardet-Biedl Syndrome Genes in the Eye. MEDICAL RESEARCH ARCHIVES 2017; 5. [PMID: 29457131 DOI: 10.18103/mra.v5i9.1526] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Upwards of 90% of individuals with Bardet-Biedl syndrome (BBS) display rod-cone dystrophy with early macular involvement. BBS is an autosomal recessive, genetically heterogeneous, pleiotropic ciliopathy for which 21 causative genes have been discovered to date. In addition to retinal degeneration, the cardinal features of BBS include obesity, cognitive impairment, renal anomalies, polydactyly, and hypogonadism. Here, we review the genes, proteins, and protein complexes involved in BBS and the BBS model organisms available for the study of retinal degeneration. We include comprehensive lists for all known BBS genes, their known phenotypes, and the model organisms available. We also review the molecular mechanisms believed to lead to retinal degeneration. We provide an overview of the mode of inheritance and describe the relationships between BBS genes and Joubert syndrome, Leber Congenital Amaurosis, Senior-Løken syndrome, and non-syndromic retinitis pigmentosa. Finally, we propose ways that new advances in technology will allow us to better understand the role of different BBS genes in retinal formation and function.
Collapse
Affiliation(s)
- Katie Weihbrecht
- Department of Pediatrics, University of Iowa; Iowa City, IA 52242, USA.,Department of Ophthalmology and Visual Sciences, University of Iowa; Iowa City, IA 52242, USA.,Stephen A. Wynn Institute for Vision Research, University of Iowa; Iowa City, IA 52242, USA
| | - Wesley A Goar
- Department of Pediatrics, University of Iowa; Iowa City, IA 52242, USA.,Department of Ophthalmology and Visual Sciences, University of Iowa; Iowa City, IA 52242, USA.,Stephen A. Wynn Institute for Vision Research, University of Iowa; Iowa City, IA 52242, USA
| | - Thomas Pak
- Department of Pediatrics, University of Iowa; Iowa City, IA 52242, USA.,Stephen A. Wynn Institute for Vision Research, University of Iowa; Iowa City, IA 52242, USA
| | - Janelle E Garrison
- Department of Pediatrics, University of Iowa; Iowa City, IA 52242, USA.,Stephen A. Wynn Institute for Vision Research, University of Iowa; Iowa City, IA 52242, USA
| | - Adam P DeLuca
- Department of Ophthalmology and Visual Sciences, University of Iowa; Iowa City, IA 52242, USA.,Stephen A. Wynn Institute for Vision Research, University of Iowa; Iowa City, IA 52242, USA
| | - Edwin M Stone
- Department of Ophthalmology and Visual Sciences, University of Iowa; Iowa City, IA 52242, USA.,Stephen A. Wynn Institute for Vision Research, University of Iowa; Iowa City, IA 52242, USA
| | - Todd E Scheetz
- Department of Ophthalmology and Visual Sciences, University of Iowa; Iowa City, IA 52242, USA.,Stephen A. Wynn Institute for Vision Research, University of Iowa; Iowa City, IA 52242, USA
| | - Val C Sheffield
- Department of Pediatrics, University of Iowa; Iowa City, IA 52242, USA.,Department of Ophthalmology and Visual Sciences, University of Iowa; Iowa City, IA 52242, USA.,Stephen A. Wynn Institute for Vision Research, University of Iowa; Iowa City, IA 52242, USA
| |
Collapse
|
24
|
Prieto-Echagüe V, Lodh S, Colman L, Bobba N, Santos L, Katsanis N, Escande C, Zaghloul NA, Badano JL. BBS4 regulates the expression and secretion of FSTL1, a protein that participates in ciliogenesis and the differentiation of 3T3-L1. Sci Rep 2017; 7:9765. [PMID: 28852127 PMCID: PMC5575278 DOI: 10.1038/s41598-017-10330-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 08/08/2017] [Indexed: 01/01/2023] Open
Abstract
Bardet-Biedl syndrome is a model ciliopathy. Although the characterization of BBS proteins has evidenced their involvement in cilia, extraciliary functions for some of these proteins are also being recognized. Importantly, understanding both cilia and cilia-independent functions of the BBS proteins is key to fully dissect the cellular basis of the syndrome. Here we characterize a functional interaction between BBS4 and the secreted protein FSTL1, a protein linked to adipogenesis and inflammation among other functions. We show that BBS4 and cilia regulate FSTL1 mRNA levels, but BBS4 also modulates FSTL1 secretion. Moreover, we show that FSTL1 is a novel regulator of ciliogenesis thus underscoring a regulatory loop between FSTL1 and cilia. Finally, our data indicate that BBS4, cilia and FSTL1 are coordinated during the differentiation of 3T3-L1 cells and that FSTL1 plays a role in this process, at least in part, by modulating ciliogenesis. Therefore, our findings are relevant to fully understand the development of BBS-associated phenotypes such as obesity.
Collapse
Affiliation(s)
- Victoria Prieto-Echagüe
- Human Molecular Genetics Laboratory, Institut Pasteur de Montevideo, Mataojo 2020, Montevideo, CP11400, Uruguay.,INDICyO Institutional Program, Institut Pasteur de Montevideo, Mataojo 2020, Montevideo, CP11400, Uruguay
| | - Sukanya Lodh
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Laura Colman
- INDICyO Institutional Program, Institut Pasteur de Montevideo, Mataojo 2020, Montevideo, CP11400, Uruguay.,Metabolic Diseases and Aging Laboratory, Institut Pasteur de Montevideo, Mataojo 2020, Montevideo, CP11400, Uruguay
| | - Natalia Bobba
- INDICyO Institutional Program, Institut Pasteur de Montevideo, Mataojo 2020, Montevideo, CP11400, Uruguay.,Metabolic Diseases and Aging Laboratory, Institut Pasteur de Montevideo, Mataojo 2020, Montevideo, CP11400, Uruguay
| | - Leonardo Santos
- INDICyO Institutional Program, Institut Pasteur de Montevideo, Mataojo 2020, Montevideo, CP11400, Uruguay.,Metabolic Diseases and Aging Laboratory, Institut Pasteur de Montevideo, Mataojo 2020, Montevideo, CP11400, Uruguay
| | - Nicholas Katsanis
- Department of Cell Biology and Center for Human Disease Modeling, Duke University Medical Center, Durham, NC, 27710, USA
| | - Carlos Escande
- INDICyO Institutional Program, Institut Pasteur de Montevideo, Mataojo 2020, Montevideo, CP11400, Uruguay.,Metabolic Diseases and Aging Laboratory, Institut Pasteur de Montevideo, Mataojo 2020, Montevideo, CP11400, Uruguay
| | - Norann A Zaghloul
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Jose L Badano
- Human Molecular Genetics Laboratory, Institut Pasteur de Montevideo, Mataojo 2020, Montevideo, CP11400, Uruguay. .,INDICyO Institutional Program, Institut Pasteur de Montevideo, Mataojo 2020, Montevideo, CP11400, Uruguay.
| |
Collapse
|
25
|
Priya S, Nampoothiri S, Sen P, Sripriya S. Bardet-Biedl syndrome: Genetics, molecular pathophysiology, and disease management. Indian J Ophthalmol 2017; 64:620-627. [PMID: 27853007 PMCID: PMC5151149 DOI: 10.4103/0301-4738.194328] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Primary cilia play a key role in sensory perception and various signaling pathways. Any defect in them leads to group of disorders called ciliopathies, and Bardet–Biedl syndrome (BBS, OMIM 209900) is one among them. The disorder is clinically and genetically heterogeneous, with various primary and secondary clinical manifestations, and shows autosomal recessive inheritance and highly prevalent in inbred/consanguineous populations. The disease mapped to at least twenty different genes (BBS1-BBS20), follow oligogenic inheritance pattern. BBS proteins localizes to the centerosome and regulates the biogenesis and functions of the cilia. In BBS, the functioning of various systemic organs (with ciliated cells) gets deranged and results in systemic manifestations. Certain components of the disease (such as obesity, diabetes, and renal problems) when noticed earlier offer a disease management benefit to the patients. However, the awareness of the disease is comparatively low and most often noticed only after severe vision loss in patients, which is usually in the first decade of the patient's age. In the current review, we have provided the recent updates retrieved from various types of scientific literature through journals, on the genetics, its molecular relevance, and the clinical outcome in BBS. The review in nutshell would provide the basic awareness of the disease that will have an impact in disease management and counseling benefits to the patients and their families.
Collapse
Affiliation(s)
- Sathya Priya
- SNONGC Department of Genetics and Molecular Biology, Kamal Nayan Institute for Research in Vision and Ophthalmology, Vision Research Foundation, Chennai, Tamil Nadu; School of Chemical and Biotechnology, SASTRA University, Thanjavur, Tamil Nadu, India
| | - Sheela Nampoothiri
- Department of Pediatric Genetics, Amrita Institute of Medical Sciences and Research Center, Cochin, Kerala, India
| | - Parveen Sen
- Department of Vitreoretina Clinic, Medical Research Foundation, Chennai, Tamil Nadu, India
| | - S Sripriya
- SNONGC Department of Genetics and Molecular Biology, Kamal Nayan Institute for Research in Vision and Ophthalmology, Vision Research Foundation, Chennai, Tamil Nadu, India
| |
Collapse
|
26
|
Abstract
Obesity, a major risk factor for the development of diabetes mellitus, cardiovascular diseases and certain types of cancer, arises from a chronic positive energy balance that is often due to unlimited access to food and an increasingly sedentary lifestyle on the background of a genetic and epigenetic vulnerability. Our understanding of the humoral and neuronal systems that mediate the control of energy homeostasis has improved dramatically in the past few decades. However, our ability to develop effective strategies to slow the current epidemic of obesity has been hampered, largely owing to the limited knowledge of the mechanisms underlying resistance to the action of metabolic hormones such as leptin and ghrelin. The development of resistance to leptin and ghrelin, hormones that are crucial for the neuroendocrine control of energy homeostasis, is a hallmark of obesity. Intensive research over the past several years has yielded tremendous progress in our understanding of the cellular pathways that disrupt the action of leptin and ghrelin. In this Review, we discuss the molecular mechanisms underpinning resistance to leptin and ghrelin and how they can be exploited as targets for pharmacological management of obesity.
Collapse
Affiliation(s)
- Huxing Cui
- Department of Pharmacology, University of Iowa, Iowa City, Iowa 52246, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242, USA
| | - Miguel López
- Department of Physiology, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain
- Centro de Investigación Biomédica en Red-Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain
| | - Kamal Rahmouni
- Department of Pharmacology, University of Iowa, Iowa City, Iowa 52246, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242, USA
| |
Collapse
|
27
|
Simple and complex retinal dystrophies are associated with profoundly different disease networks. Sci Rep 2017; 7:41835. [PMID: 28139756 PMCID: PMC5282568 DOI: 10.1038/srep41835] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 12/28/2016] [Indexed: 12/20/2022] Open
Abstract
Retinopathies are a group of monogenetic or complex retinal diseases associated with high unmet medical need. Monogenic disorders are caused by rare genetic variation and usually arise early in life. Other diseases, such as age-related macular degeneration (AMD), develop late in life and are considered to be of complex origin as they develop from a combination of genetic, ageing, environmental and lifestyle risk factors. Here, we contrast the underlying disease networks and pathological mechanisms of monogenic as opposed to complex retinopathies, using AMD as an example of the latter. We show that, surprisingly, genes associated with the different forms of retinopathies in general do not overlap despite their overlapping retinal phenotypes. Further, AMD risk genes participate in multiple networks with interaction partners that link to different ubiquitous pathways affecting general tissue integrity and homeostasis. Thus AMD most likely represents an endophenotype with differing underlying pathogenesis in different subjects. Localising these pathomechanisms and processes within and across different retinal anatomical compartments provides a novel representation of AMD that may be extended to complex disease in general. This approach may generate improved treatment options that target multiple processes with the aim of restoring tissue homeostasis and maintaining vision.
Collapse
|
28
|
Wauman J, Zabeau L, Tavernier J. The Leptin Receptor Complex: Heavier Than Expected? Front Endocrinol (Lausanne) 2017; 8:30. [PMID: 28270795 PMCID: PMC5318964 DOI: 10.3389/fendo.2017.00030] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/01/2017] [Indexed: 12/31/2022] Open
Abstract
Under normal physiological conditions, leptin and the leptin receptor (ObR) regulate the body weight by balancing food intake and energy expenditure. However, this adipocyte-derived hormone also directs peripheral processes, including immunity, reproduction, and bone metabolism. Leptin, therefore, can act as a metabolic switch connecting the body's nutritional status to high energy consuming processes. We provide an extensive overview of current structural insights on the leptin-ObR interface and ObR activation, coupling to signaling pathways and their negative regulation, and leptin functioning under normal and pathophysiological conditions (obesity, autoimmunity, cancer, … ). We also discuss possible cross-talk with other receptor systems on the receptor (extracellular) and signaling cascade (intracellular) levels.
Collapse
Affiliation(s)
- Joris Wauman
- Cytokine Receptor Laboratory, Faculty of Medicine and Health Sciences, Department of Biochemistry, Ghent University, Ghent, Belgium
- VIB Medical Biotechnology Center, VIB, Ghent, Belgium
| | - Lennart Zabeau
- Cytokine Receptor Laboratory, Faculty of Medicine and Health Sciences, Department of Biochemistry, Ghent University, Ghent, Belgium
- VIB Medical Biotechnology Center, VIB, Ghent, Belgium
| | - Jan Tavernier
- Cytokine Receptor Laboratory, Faculty of Medicine and Health Sciences, Department of Biochemistry, Ghent University, Ghent, Belgium
- VIB Medical Biotechnology Center, VIB, Ghent, Belgium
- *Correspondence: Jan Tavernier,
| |
Collapse
|
29
|
Abstract
Primary cilia are small, antenna-like structures that detect mechanical and chemical cues and transduce extracellular signals. While mammalian primary cilia were first reported in the late 1800s, scientific interest in these sensory organelles has burgeoned since the beginning of the twenty-first century with recognition that primary cilia are essential to human health. Among the most common clinical manifestations of ciliary dysfunction are renal cysts. The molecular mechanisms underlying renal cystogenesis are complex, involving multiple aberrant cellular processes and signaling pathways, while initiating molecular events remain undefined. Autosomal Dominant Polycystic Kidney Disease is the most common renal cystic disease, caused by disruption of polycystin-1 and polycystin-2 transmembrane proteins, which evidence suggests must localize to primary cilia for proper function. To understand how the absence of these proteins in primary cilia may be remediated, we review intracellular trafficking of polycystins to the primary cilium. We also examine the controversial mechanisms by which primary cilia transduce flow-mediated mechanical stress into intracellular calcium. Further, to better understand ciliary function in the kidney, we highlight the LKB1/AMPK, Wnt, and Hedgehog developmental signaling pathways mediated by primary cilia and misregulated in renal cystic disease.
Collapse
|
30
|
Lutz TA. The brain needs interleukin-6 (IL-6) to maintain a "healthy" energy balance. Focus on "IL-6 ameliorates defective leptin sensitivity in DIO ventromedial hypothalamic nucleus neurons". Am J Physiol Regul Integr Comp Physiol 2016; 311:R989-R991. [PMID: 27733389 DOI: 10.1152/ajpregu.00426.2016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 10/07/2016] [Accepted: 10/07/2016] [Indexed: 11/22/2022]
Affiliation(s)
- Thomas A Lutz
- Institute of Veterinary Physiology and Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| |
Collapse
|
31
|
Jacobs DT, Silva LM, Allard BA, Schonfeld MP, Chatterjee A, Talbott GC, Beier DR, Tran PV. Dysfunction of intraflagellar transport-A causes hyperphagia-induced obesity and metabolic syndrome. Dis Model Mech 2016; 9:789-98. [PMID: 27482817 PMCID: PMC4958314 DOI: 10.1242/dmm.025791] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 04/29/2016] [Indexed: 12/15/2022] Open
Abstract
Primary cilia extend from the plasma membrane of most vertebrate cells and mediate signaling pathways. Ciliary dysfunction underlies ciliopathies, which are genetic syndromes that manifest multiple clinical features, including renal cystic disease and obesity. THM1 (also termed TTC21B or IFT139) encodes a component of the intraflagellar transport-A complex and mutations in THM1 have been identified in 5% of individuals with ciliopathies. Consistent with this, deletion of murine Thm1 during late embryonic development results in cystic kidney disease. Here, we report that deletion of murine Thm1 during adulthood results in obesity, diabetes, hypertension and fatty liver disease, with gender differences in susceptibility to weight gain and metabolic dysfunction. Pair-feeding of Thm1 conditional knock-out mice relative to control littermates prevented the obesity and related disorders, indicating that hyperphagia caused the obese phenotype. Thm1 ablation resulted in increased localization of adenylyl cyclase III in primary cilia that were shortened, with bulbous distal tips on neurons of the hypothalamic arcuate nucleus, an integrative center for signals that regulate feeding and activity. In pre-obese Thm1 conditional knock-out mice, expression of anorexogenic pro-opiomelanocortin (Pomc) was decreased by 50% in the arcuate nucleus, which likely caused the hyperphagia. Fasting of Thm1 conditional knock-out mice did not alter Pomc nor orexogenic agouti-related neuropeptide (Agrp) expression, suggesting impaired sensing of changes in peripheral signals. Together, these data indicate that the Thm1-mutant ciliary defect diminishes sensitivity to feeding signals, which alters appetite regulation and leads to hyperphagia, obesity and metabolic disease. Summary: Disruption of the IFT-A complex gene, Thm1, in adult mice misregulates response to feeding signals, altering appetite regulation and resulting in obesity through hyperphagia.
Collapse
Affiliation(s)
- Damon T Jacobs
- Department of Anatomy and Cell Biology and The Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Luciane M Silva
- Department of Anatomy and Cell Biology and The Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Bailey A Allard
- Department of Anatomy and Cell Biology and The Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Michael P Schonfeld
- Department of Anatomy and Cell Biology and The Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Anindita Chatterjee
- Department of Anatomy and Cell Biology and The Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - George C Talbott
- Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - David R Beier
- Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98105, USA
| | - Pamela V Tran
- Department of Anatomy and Cell Biology and The Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
32
|
|
33
|
Guo DF, Cui H, Zhang Q, Morgan DA, Thedens DR, Nishimura D, Grobe JL, Sheffield VC, Rahmouni K. The BBSome Controls Energy Homeostasis by Mediating the Transport of the Leptin Receptor to the Plasma Membrane. PLoS Genet 2016; 12:e1005890. [PMID: 26926121 PMCID: PMC4771807 DOI: 10.1371/journal.pgen.1005890] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Accepted: 01/31/2016] [Indexed: 02/05/2023] Open
Abstract
Bardet-Biedl syndrome (BBS) is a highly pleiotropic autosomal recessive disorder associated with a wide range of phenotypes including obesity. However, the underlying mechanism remains unclear. Here, we show that neuronal BBSome is a critical determinant of energy balance through its role in the regulation of the trafficking of the long signaling form of the leptin receptor (LRb). Targeted disruption of the BBSome by deleting the Bbs1 gene from the nervous system causes obesity in mice, and this phenotype is reproduced by ablation of the Bbs1 gene selectively in the LRb-expressing cells, but not from adipocytes. Obesity developed as a consequence of both increased food intake and decreased energy expenditure in mice lacking the Bbs1 gene in LRb-expressing cells. Strikingly, the well-known role of BBS proteins in the regulation of ciliary formation and function is unlikely to account for the obesogenic effect of BBS1 loss as disruption of the intraflagellar transport (IFT) machinery required for ciliogenesis by deleting the Ift88 gene in LRb-expressing cells caused a marginal increase in body weight and adiposity. Instead, we demonstrate that silencing BBS proteins, but not IFT88, impair the trafficking of the LRb to the plasma membrane leading to central leptin resistance in a manner independent of obesity. Our data also demonstrate that postnatal deletion of the Bbs1 gene in the mediobasal hypothalamus can cause obesity in mice, arguing against an early neurodevelopmental origin of obesity in BBS. Our results depict a novel mechanism underlying energy imbalance and obesity in BBS with potential implications in common forms of human obesity. The brain plays an important role in maintaining energy homeostasis. The hormone leptin is a critical afferent signal in metabolic homeostasis through its action in the brain. Here we show that neuronal Bardet-Biedl syndrome (BBS) proteins, encoded by genes that cause obesity when mutated, govern energy homeostasis through the control of cell surface expression of the leptin receptor. Selective disruption of BBS proteins causes obesity in mice and impairs the transport of the leptin receptor to the plasma membrane leading to leptin resistance in a manner independent of obesity. These results establish BBS proteins as a fundamental mechanism underlying transport of the leptin receptor and explain why BBS patients develop obesity.
Collapse
Affiliation(s)
- Deng-Fu Guo
- Department of Pharmacology, University of Iowa, Iowa City, Iowa, United States of America
| | - Huxing Cui
- Department of Pharmacology, University of Iowa, Iowa City, Iowa, United States of America
| | - Qihong Zhang
- Department of Pediatrics, University of Iowa, Iowa City, Iowa, United States of America
| | - Donald A Morgan
- Department of Pharmacology, University of Iowa, Iowa City, Iowa, United States of America
| | - Daniel R Thedens
- Department of Radiology, University of Iowa, Iowa City, Iowa, United States of America
| | - Darryl Nishimura
- Department of Pediatrics, University of Iowa, Iowa City, Iowa, United States of America
| | - Justin L Grobe
- Department of Pharmacology, University of Iowa, Iowa City, Iowa, United States of America.,Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa, United States of America
| | - Val C Sheffield
- Department of Pediatrics, University of Iowa, Iowa City, Iowa, United States of America
| | - Kamal Rahmouni
- Department of Pharmacology, University of Iowa, Iowa City, Iowa, United States of America.,Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa, United States of America.,Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States of America
| |
Collapse
|
34
|
Mason K, Page L, Balikcioglu PG. Screening for hormonal, monogenic, and syndromic disorders in obese infants and children. Pediatr Ann 2014; 43:e218-24. [PMID: 25198446 PMCID: PMC4369917 DOI: 10.3928/00904481-20140825-08] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The prevalence of pediatric obesity in the United States is nearly 17%. Most cases are "exogenous", resulting from excess energy intake relative to energy expenditure over a prolonged period of time. However, some cases of obesity are "endogenous", associated with hormonal, genetic, or syndromic disorders such as hypothyroidism, Cushing's syndrome, growth hormone deficiency, defective leptin signaling, mutations in the melanocortin 4 receptor, and Prader-Willi and Bardet-Biedl syndromes. This article reviews the hormonal, monogenic, and syndromic causes of childhood obesity and identifies critical features that distinguish "endogenous" obesity disorders from the more common exogenous obesity. Findings that raise suspicion for endogenous obesity include onset in infancy, lack of satiety, poor linear growth, dysmorphic features, and cognitive dysfunction. Selection and interpretation of appropriate laboratory tests and indications for subspecialist referral are also discussed.
Collapse
|
35
|
Xie YA, Lee W, Cai C, Gambin T, Nõupuu K, Sujirakul T, Ayuso C, Jhangiani S, Muzny D, Boerwinkle E, Gibbs R, Greenstein VC, Lupski JR, Tsang SH, Allikmets R. New syndrome with retinitis pigmentosa is caused by nonsense mutations in retinol dehydrogenase RDH11. Hum Mol Genet 2014; 23:5774-80. [PMID: 24916380 DOI: 10.1093/hmg/ddu291] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Retinitis pigmentosa (RP), a genetically heterogeneous group of retinopathies that occur in both non-syndromic and syndromic forms, is caused by mutations in ∼100 genes. Although recent advances in next-generation sequencing have aided in the discovery of novel RP genes, a number of the underlying contributing genes and loci remain to be identified. We investigated three siblings, born to asymptomatic parents of Italian-American descent, who each presented with atypical RP with systemic features, including facial dysmorphologies, psychomotor developmental delays recognized since early childhood, learning disabilities and short stature. RP-associated ophthalmological findings included salt-and-pepper retinopathy, attenuation of the arterioles and generalized rod-cone dysfunction as determined by almost extinguished electroretinogram in 2 of 3 siblings. Atypical for RP features included mottled macula at an early age and peripapillary sparing of the retinal pigment epithelium. Whole-exome sequencing data, queried under a recessive model of inheritance, identified compound heterozygous stop mutations, c.C199T:p.R67* and c.C322T:p.R108*, in the retinol dehydrogenase 11 (RDH11) gene, resulting in a non-functional protein, in all affected children. In summary, deleterious mutations in RDH11, an important enzyme for vision-related and systemic retinoic acid metabolism, cause a new syndrome with RP.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Carmen Ayuso
- Department of Genetics, Instituto de Investigacion Sanitaria-University Hospital Fundacion Jimenez Diaz (IIS-FJD), Madrid, 28040 Spain and Centro de Investigacion Biomedica en Red (CIBER) de Enfermedades Raras, ISCIII, Madrid, 28040 Spain
| | | | - Donna Muzny
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Richard Gibbs
- Department of Molecular and Human Genetics, Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - James R Lupski
- Department of Molecular and Human Genetics, Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Stephen H Tsang
- Department of Ophthalmology, Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Rando Allikmets
- Department of Ophthalmology, Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA,
| |
Collapse
|
36
|
Acs P, Bauer PO, Mayer B, Bera T, Macallister R, Mezey E, Pastan I. A novel form of ciliopathy underlies hyperphagia and obesity in Ankrd26 knockout mice. Brain Struct Funct 2014; 220:1511-28. [PMID: 24633808 DOI: 10.1007/s00429-014-0741-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 02/19/2014] [Indexed: 02/06/2023]
Abstract
Human ciliopathies are genetic disorders caused by mutations in genes responsible for the formation and function of primary cilia. Some are associated with hyperphagia and obesity (e.g., Bardet-Biedl Syndrome, Alström Syndrome), but the mechanisms underlying these problems are not fully understood. The human gene ANKRD26 is located on 10p12, a locus that is associated with some forms of hereditary obesity. Previously, we reported that disruption of this gene causes hyperphagia, obesity and gigantism in mice. In the present study, we looked for the mechanisms that induce hyperphagia in the Ankrd26-/- mice and found defects in primary cilia in regions of the central nervous system that control appetite and energy homeostasis.
Collapse
Affiliation(s)
- Peter Acs
- Laboratory of Molecular Biology, National Cancer Institute, NIH, Bethesda, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Heymsfield SB, Avena NM, Baier L, Brantley P, Bray GA, Burnett LC, Butler MG, Driscoll DJ, Egli D, Elmquist J, Forster JL, Goldstone AP, Gourash LM, Greenway FL, Han JC, Kane JG, Leibel RL, Loos RJ, Scheimann AO, Roth CL, Seeley RJ, Sheffield V, Tauber M, Vaisse C, Wang L, Waterland RA, Wevrick R, Yanovski JA, Zinn AR. Hyperphagia: current concepts and future directions proceedings of the 2nd international conference on hyperphagia. Obesity (Silver Spring) 2014; 22 Suppl 1:S1-S17. [PMID: 24574081 PMCID: PMC4159941 DOI: 10.1002/oby.20646] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 10/11/2013] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Hyperphagia is a central feature of inherited disorders (e.g., Prader-Willi Syndrome) in which obesity is a primary phenotypic component. Hyperphagia may also contribute to obesity as observed in the general population, thus raising the potential importance of common underlying mechanisms and treatments. Substantial gaps in understanding the molecular basis of inherited hyperphagia syndromes are present as are a lack of mechanistic of mechanistic targets that can serve as a basis for pharmacologic and behavioral treatments. DESIGN AND METHODS International conference with 28 experts, including scientists and caregivers, providing presentations, panel discussions, and debates. RESULTS The reviewed collective research and clinical experience provides a critical body of new and novel information on hyperphagia at levels ranging from molecular to population. Gaps in understanding and tools needed for additional research were identified. CONCLUSIONS This report documents the full scope of important topics reviewed at a comprehensive international meeting devoted to the topic of hyperphagia and identifies key areas for future funding and research.
Collapse
Affiliation(s)
- Steven B. Heymsfield
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA
| | - Nicole M. Avena
- Department of Psychiatry, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Leslie Baier
- Diabetes Molecular Genetics Section, Phoenix Epidemiology and Clinical Research Branch, NIDDK, NIH, Phoenix, Arizona, USA
| | - Phillip Brantley
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA
| | - George A. Bray
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA
| | - Lisa C. Burnett
- College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | | | - Daniel J. Driscoll
- Division of Genetics and Metabolism, Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Dieter Egli
- College of Physicians and Surgeons, Columbia University, New York, New York, USA
- New York Stem Cell Foundation, New York, New York, USA
| | | | | | - Anthony P. Goldstone
- Metabolic & Molecular Imaging Group, MRC Clinical Sciences Centre, Imperial College London, UK
| | | | - Frank L. Greenway
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA
| | - Joan C. Han
- Section on Growth and Obesity, National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| | - James G. Kane
- Prader-Willi Syndrome Association (USA), Sarasota, Florida, USA
| | - Rudolph L. Leibel
- College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Ruth J.F. Loos
- The Genetics of Obesity and Related Metabolic Traits Program, The Charles Bronfman Institute for Personalized Medicine, The Mindich Child Health and Development Institute, The Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ann O. Scheimann
- Division of Pediatric Gastroenterology, Nutrition and Hepatology at Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Christian L. Roth
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Randy J. Seeley
- Center of Excellence in Obesity and Diabetes, University of Cincinnati, Cincinnati, Ohio, USA
| | - Val Sheffield
- Pediatrics and Medical Genetics, University of Iowa College of Medicine, Iowa City, Iowa, USA
| | - Maïthé Tauber
- Department of Endocrinology, Hôpital des Enfants and Paul Sabatier Université, Toulouse, France
| | - Christian Vaisse
- University of California, San Francisco, School of Medicine, San Francisco, California, USA
| | - Liheng Wang
- College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Robert A. Waterland
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics and Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Rachel Wevrick
- Department of Medical Genetics, University of Alberta, Edmonton, Canada
| | - Jack A. Yanovski
- Section on Growth and Obesity, National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| | - Andrew R. Zinn
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
38
|
Lee EB, Mattson MP. The neuropathology of obesity: insights from human disease. Acta Neuropathol 2014; 127:3-28. [PMID: 24096619 DOI: 10.1007/s00401-013-1190-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 09/27/2013] [Accepted: 09/28/2013] [Indexed: 02/06/2023]
Abstract
Obesity, a pathologic state defined by excess adipose tissue, is a significant public health problem as it affects a large proportion of individuals and is linked with increased risk for numerous chronic diseases. Obesity is the result of fundamental changes associated with modern society including overnutrition and sedentary lifestyles. Proper energy homeostasis is dependent on normal brain function as the master metabolic regulator, which integrates peripheral signals, modulates autonomic outflow and controls feeding behavior. Therefore, many human brain diseases are associated with obesity. This review explores the neuropathology of obesity by examining brain diseases which either cause or are influenced by obesity. First, several genetic and acquired brain diseases are discussed as a means to understand the central regulation of peripheral metabolism. These diseases range from monogenetic causes of obesity (leptin deficiency, MC4R deficiency, Bardet-Biedl syndrome and others) to complex neurodevelopmental disorders (Prader-Willi syndrome and Sim1 deficiency) and neurodegenerative conditions (frontotemporal dementia and Gourmand's syndrome) and serve to highlight the central regulatory mechanisms which have evolved to maintain energy homeostasis. Next, to examine the effect of obesity on the brain, chronic neuropathologic conditions (epilepsy, multiple sclerosis and Alzheimer's disease) are discussed as examples of obesity leading to maladaptive processes which exacerbate chronic disease. Thus, obesity is associated with multiple pathways including abnormal metabolism, altered hormonal signaling and increased inflammation which act in concert to promote downstream neuropathology. Finally, the effect of anti-obesity interventions is discussed in terms of brain structure and function. Together, understanding human diseases and anti-obesity interventions leads to insights into the bidirectional interaction between peripheral metabolism and central brain function, highlighting the need for continued clinicopathologic and mechanistic studies of the neuropathology of obesity.
Collapse
|
39
|
Sherafat-Kazemzadeh R, Ivey L, Kahn SR, Sapp JC, Hicks MD, Kim RC, Krause AJ, Shomaker LB, Biesecker LG, Han JC, Yanovski JA. Hyperphagia among patients with Bardet-Biedl syndrome. Pediatr Obes 2013; 8:e64-7. [PMID: 23776152 PMCID: PMC3901011 DOI: 10.1111/j.2047-6310.2013.00182.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Revised: 04/30/2013] [Accepted: 05/06/2013] [Indexed: 11/28/2022]
Abstract
BACKGROUND The importance of hyperphagia as a cause for energy imbalance in humans with Bardet-Biedl syndrome (BBS) has not been established. We therefore compared hyperphagic symptoms in patients with BBS vs. controls. METHODS We studied 13 patients with BBS and 23 non-syndromic controls with similar age, sex and body mass index (BMI) z-score. A 13-item hyperphagia questionnaire was completed by patients' parents/guardians. RESULTS Total hyperphagia questionnaire score was higher in BBS than controls (27.6 ± 9.0 vs. 19.1 ± 7.9, P = 0.005). Behaviour and drive subscales were higher for BBS than controls (12.5 ± 4.1 vs. 7.8 ± 3.2, P = 0.001, and 11.2 ± 4.1 vs. 8.3 ± 3.8, P = 0.04, respectively); severity was not significantly different between groups (3.8 ± 1.5 vs. 3.0 ± 1.3, P = 0.072). After adjustment for demographic variables and BMI z-score, total and behaviour subscale scores remained significantly different between groups, suggesting food-seeking activity, rather than preoccupation with food may be the main hyperphagic feature among patients with BBS. CONCLUSION Appetite dysregulation may contribute to obesity in BBS.
Collapse
Affiliation(s)
- Roya Sherafat-Kazemzadeh
- Section on Growth and Obesity (SGO), Program in Developmental Endocrinology and Genetics (PDEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), DHHS, Bethesda, MD 20892, USA
| | - Lauren Ivey
- Human Development Section, Genetic Disease Research Branch, National Human Genome Research Institute, NIH
| | - Stephanie R. Kahn
- Section on Growth and Obesity (SGO), Program in Developmental Endocrinology and Genetics (PDEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), DHHS, Bethesda, MD 20892, USA
| | - Julie C. Sapp
- Human Development Section, Genetic Disease Research Branch, National Human Genome Research Institute, NIH
| | - Melanie D. Hicks
- Section on Growth and Obesity (SGO), Program in Developmental Endocrinology and Genetics (PDEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), DHHS, Bethesda, MD 20892, USA
- Unit on Metabolism and Neuroendocrinology, SGO, PDEGEN, NICHD, NIH, DHHS, Bethesda, MD 20892, USA
| | - Rachel C. Kim
- Section on Growth and Obesity (SGO), Program in Developmental Endocrinology and Genetics (PDEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), DHHS, Bethesda, MD 20892, USA
- Unit on Metabolism and Neuroendocrinology, SGO, PDEGEN, NICHD, NIH, DHHS, Bethesda, MD 20892, USA
| | - Amanda J. Krause
- Section on Growth and Obesity (SGO), Program in Developmental Endocrinology and Genetics (PDEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), DHHS, Bethesda, MD 20892, USA
| | - Lauren B. Shomaker
- Section on Growth and Obesity (SGO), Program in Developmental Endocrinology and Genetics (PDEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), DHHS, Bethesda, MD 20892, USA
| | - Leslie G. Biesecker
- Human Development Section, Genetic Disease Research Branch, National Human Genome Research Institute, NIH
| | - Joan C. Han
- Section on Growth and Obesity (SGO), Program in Developmental Endocrinology and Genetics (PDEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), DHHS, Bethesda, MD 20892, USA
- Unit on Metabolism and Neuroendocrinology, SGO, PDEGEN, NICHD, NIH, DHHS, Bethesda, MD 20892, USA
| | - Jack A. Yanovski
- Section on Growth and Obesity (SGO), Program in Developmental Endocrinology and Genetics (PDEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), DHHS, Bethesda, MD 20892, USA
| |
Collapse
|
40
|
Gerhard GS, Chu X, Wood GC, Gerhard GM, Benotti P, Petrick AT, Gabrielsen J, Strodel WE, Still CD, Argyropoulos G. Next-generation sequence analysis of genes associated with obesity and nonalcoholic fatty liver disease-related cirrhosis in extreme obesity. Hum Hered 2013; 75:144-51. [PMID: 24081230 DOI: 10.1159/000351719] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES Genome-wide association studies (GWAS) have led to the identification of single nucleotide polymorphisms in or near several loci that are associated with the risk of obesity and nonalcoholic fatty liver disease (NAFLD). We hypothesized that missense variants in GWAS and related candidate genes may underlie cases of extreme obesity and NAFLD-related cirrhosis, an extreme manifestation of NAFLD. METHODS We performed whole-exome sequencing on 6 Caucasian patients with extreme obesity [mean body mass index (BMI) 84.4] and 4 obese Caucasian patients (mean BMI 57.0) with NAFLD-related cirrhosis. RESULTS Sequence analysis was performed on 24 replicated GWAS and selected candidate obesity genes and 5 loci associated with NAFLD. No missense variants were identified in 19 of the 29 genes analyzed, although all patients carried at least 2 missense variants in the remaining genes without excess homozygosity. One patient with extreme obesity carried 2 novel damaging mutations in BBS1 and was homozygous for benign and damaging MC3R variants. In addition, 1 patient with NAFLD-related cirrhosis was compound heterozygous for rare damaging mutations in PNPLA3. CONCLUSIONS These results indicate that analyzing candidate loci previously identified by GWAS analyses using whole-exome sequencing is an effective strategy to identify potentially causative missense variants underlying extreme obesity and NAFLD-related cirrhosis.
Collapse
Affiliation(s)
- Glenn S Gerhard
- Geisinger Obesity Research Institute, Geisinger Clinic, Danville, Pa., USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Bay SN, Caspary T. What are those cilia doing in the neural tube? Cilia 2012; 1:19. [PMID: 23351466 PMCID: PMC3556023 DOI: 10.1186/2046-2530-1-19] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 06/29/2012] [Indexed: 12/31/2022] Open
Abstract
Primary cilia are present on almost all vertebrate cells, and they have diverse functions in distinct tissues. Cilia are important for sensation in multiple capacities in contexts as different as the retina, kidney, and inner ear. In addition to these roles, cilia play a critical part in various developmental processes. Of particular importance is the development of the neural tube, where cilia are essential for the transduction of the Sonic Hedgehog (Shh) signaling pathway that specifies neuronal cell fates. This relationship is well established and is the most recognizable function for cilia in the neural tube, but it may be part of a larger picture. Here, we discuss the links between cilia and Shh signaling, as well as suggesting additional roles for cilia, and mechanisms for their placement, in the neural tube.
Collapse
Affiliation(s)
- Sarah N Bay
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | | |
Collapse
|
42
|
Abstract
Bardet-Biedl syndrome (BBS) is a rare autosomal recessive ciliopathy characterised by retinal dystrophy, obesity, post-axial polydactyly, renal dysfunction, learning difficulties and hypogonadism. Many associated minor features can be helpful in making a diagnosis and are important in the clinical management of BBS. The diagnosis is based on clinical findings and can be confirmed by sequencing of known disease-causing genes in 80% of patients. BBS genes encode proteins that localise to the cilia and basal body and are involved in cilia biogenesis and function. Mutations lead to defective cilia accounting in part for the pleiotropic effects observed in BBS. We provide an overview of BBS including the clinical findings, current understanding of cilia biology, and a practical approach to diagnosis, genetic counselling and up-to-date management.
Collapse
Affiliation(s)
- Elizabeth Forsythe
- Molecular Medicine Unit, Institute of Child Health, University College London, London, UK
| | | |
Collapse
|
43
|
Saqui-Salces M, Dowdle WE, Reiter JF, Merchant JL. A high-fat diet regulates gastrin and acid secretion through primary cilia. FASEB J 2012; 26:3127-39. [PMID: 22516298 DOI: 10.1096/fj.11-197426] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The role of primary cilia in the gastrointestinal tract has not been examined. Here we report the presence of primary cilia on gastric endocrine cells producing gastrin, ghrelin, and somatostatin (Sst), hormones regulated by food intake. During eating, cilia in the gastric antrum decreased, whereas gastric acid and circulating gastrin increased. Mice fed high-fat chow showed a delayed decrease in antral cilia, increased plasma gastrin, and gastric acidity. Mice fed high-fat chow for 3 wk showed lower cilia numbers and acid but higher gastrin levels than mice fed a standard diet, suggesting that fat affects gastric physiology. Ex vivo experiments showed that cilia in the corpus responded to acid and distension, whereas cilia in the antrum responded to food. To analyze the role of gastric cilia, we conditionally deleted the intraflagellar transport protein Ift88 (Ift88(-/fl)). In fed Ift88(-/fl) mice, gastrin levels were higher, and gastric acidity was lower. Moreover, gastrin and Sst gene expression did not change in response to food as in controls. At 8 mo, Ift88(-/fl) mice developed foveolar hyperplasia, hypergastrinemia, and hypochlorhydria associated with endocrine dysfunction. Our results show that components of food (fat) are sensed by antral cilia on endocrine cells, which modulates gastrin secretion and gastric acidity.
Collapse
Affiliation(s)
- Milena Saqui-Salces
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | | |
Collapse
|
44
|
D'Angelo CS, Koiffmann CP. Copy number variants in obesity-related syndromes: review and perspectives on novel molecular approaches. J Obes 2012; 2012:845480. [PMID: 23316347 PMCID: PMC3534325 DOI: 10.1155/2012/845480] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 10/09/2012] [Indexed: 02/07/2023] Open
Abstract
In recent decades, obesity has reached epidemic proportions worldwide and became a major concern in public health. Despite heritability estimates of 40 to 70% and the long-recognized genetic basis of obesity in a number of rare cases, the list of common obesity susceptibility variants by the currently published genome-wide association studies (GWASs) only explain a small proportion of the individual variation in risk of obesity. It was not until very recently that GWASs of copy number variants (CNVs) in individuals with extreme phenotypes reported a number of large and rare CNVs conferring high risk to obesity, and specifically deletions on chromosome 16p11.2. In this paper, we comment on the recent advances in the field of genetics of obesity with an emphasis on the genes and genomic regions implicated in highly penetrant forms of obesity associated with developmental disorders. Array genomic hybridization in this patient population has afforded discovery opportunities for CNVs that have not previously been detectable. This information can be used to generate new diagnostic arrays and sequencing platforms, which will likely enhance detection of known genetic conditions with the potential to elucidate new disease genes and ultimately help in developing a next-generation sequencing protocol relevant to clinical practice.
Collapse
Affiliation(s)
- Carla Sustek D'Angelo
- Human Genome and Stem Cell Center, Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of Sao Paulo, 277 Rua do Matao, Rooms 204 and 209, 05508-090 Sao Paulo, SP, Brazil.
| | | |
Collapse
|
45
|
Bingham NC, Rose SR, Inge TH. Bariatric surgery in hypothalamic obesity. Front Endocrinol (Lausanne) 2012; 3:23. [PMID: 22649412 PMCID: PMC3355900 DOI: 10.3389/fendo.2012.00023] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Accepted: 01/30/2012] [Indexed: 12/21/2022] Open
Abstract
Craniopharyngiomas (CP) are epithelial neoplasms generally found in the area of the pituitary and hypothalamus. Despite benign histology, these tumors and/or their treatment often result in significant, debilitating disorders of endocrine, neurological, behavioral, and metabolic systems. Severe obesity is observed in a high percentage of patients with CP resulting in significant comorbidities and negatively impacting quality of life. Obesity occurs as a result of hypothalamic damage and disruption of normal homeostatic mechanisms regulating energy balance. Such pathological weight gain, termed hypothalamic obesity (HyOb), is often severe and refractory to therapy. Unfortunately, neither lifestyle intervention nor pharmacotherapy has proven effective in the treatment of HyOb. Given the limited choices and poor results of these treatments, several groups have examined bariatric surgery as a treatment alternative for patients with CP-HyOb. While a large body of evidence exists supporting the use of bariatric surgery in the treatment of exogenous obesity and its comorbidities, its role in the treatment of HyOb has yet to be defined. To date, the existing literature on bariatric surgery in CP-HyOb is largely limited to case reports and series with short term follow-up. Here we review the current reports on the use of bariatric surgery in the treatment of CP-HyOb. We also compare these results to those reported for other populations of HyOb, including Prader-Willi Syndrome, Bardet-Biedl syndrome, and hypothalamic melanocortin signaling defects. While initial reports of bariatric surgery in CP-HyOb are promising, their limited scope makes it difficult to draw any substantial conclusions as to the long term safety and efficacy of bariatric surgery in CP-HyOb. There continues to be a need for more robust, controlled, prospective studies with long term follow-up in order to better define the role of bariatric surgery in the treatment of HyOb.
Collapse
Affiliation(s)
- Nathan C Bingham
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine Cincinnati, OH, USA
| | | | | |
Collapse
|