1
|
Breusegem SY, Houghton J, Romero-Bueno R, Fragoso-Luna A, Kentistou KA, Ong KK, Janssen AFJ, Bright NA, Riedel CG, Perry JRB, Askjaer P, Larrieu D. A multiparametric anti-aging CRISPR screen uncovers a role for BAF in protein synthesis regulation. Nat Commun 2025; 16:1681. [PMID: 39956852 PMCID: PMC11830792 DOI: 10.1038/s41467-025-56916-5] [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] [Academic Contribution Register] [Received: 07/31/2023] [Accepted: 01/28/2025] [Indexed: 02/18/2025] Open
Abstract
Progeria syndromes are very rare, incurable premature aging conditions recapitulating most aging features. Here, we report a whole genome, multiparametric CRISPR screen, identifying 43 genes that can rescue multiple cellular phenotypes associated with progeria. We implement the screen in fibroblasts from Néstor-Guillermo Progeria Syndrome male patients, carrying a homozygous A12T mutation in BAF. The hits are enriched for genes involved in protein synthesis, protein and RNA transport and osteoclast formation and are validated in a whole-organism Caenorhabditis elegans model. We further confirm that BAF A12T can disrupt protein synthesis rate and fidelity, which could contribute to premature aging in patients. This work highlights the power of multiparametric genome-wide suppressor screens to identify genes enhancing cellular resilience in premature aging and provide insights into the biology underlying progeria-associated cellular dysfunction.
Collapse
Affiliation(s)
- Sophia Y Breusegem
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Keith Peters Building, Hills Road, Cambridge, UK
- Sophia Y. Breusegem: MRC toxicology Unit, University of Cambridge, Tennis Court Road, Cambridge, UK
| | - Jack Houghton
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Keith Peters Building, Hills Road, Cambridge, UK
- Jack Houghton: Imperial College London, Exhibition Road, South Kensington, London, UK
| | - Raquel Romero-Bueno
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas-Universidad Pablo de Olavide-Junta de Andalucía, Seville, Spain
| | - Adrián Fragoso-Luna
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas-Universidad Pablo de Olavide-Junta de Andalucía, Seville, Spain
| | - Katherine A Kentistou
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Ken K Ong
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Anne F J Janssen
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Keith Peters Building, Hills Road, Cambridge, UK
- Anne F. J. Janssen: Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, Nijmegen, The Netherlands
| | - Nicholas A Bright
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Keith Peters Building, Hills Road, Cambridge, UK
| | | | - John R B Perry
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
- Metabolic Research Laboratory, Wellcome-MRC Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Peter Askjaer
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas-Universidad Pablo de Olavide-Junta de Andalucía, Seville, Spain
| | - Delphine Larrieu
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Keith Peters Building, Hills Road, Cambridge, UK.
- Delphine Larrieu: Altos Labs, Cambridge Institute of Science, Cambridge, UK.
| |
Collapse
|
2
|
Huang X, Dong G, Fan H, Zhou W, Huang G, Guan D, Zhang D, Wei F. The genome of African manatee Trichechus senegalensis reveals secondary adaptation to the aquatic environment. iScience 2024; 27:110394. [PMID: 39092175 PMCID: PMC11292518 DOI: 10.1016/j.isci.2024.110394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/26/2024] [Revised: 05/26/2024] [Accepted: 06/25/2024] [Indexed: 08/04/2024] Open
Abstract
Sirenians exhibit unique aquatic adaptations, showcasing both convergent adaptive features shared with cetaceans and unique characteristics such as cold sensitivity and dense bones. Here, we report a chromosome-level genome of the African manatee (Trichechus senegalensis) with high continuity, completeness, and accuracy. We found that genes associated with osteopetrosis have undergone positive selection (CSF1R and LRRK1) or pseudogenized (FAM111A and IGSF23) in the African manatee, potentially contributing to the dense bone formation. The loss of KCNK18 may have increased their sensitivity to cold water temperatures. Moreover, we identified convergent evolutionary signatures in 392 genes among fully aquatic mammals, primarily enriched in skin or skeletal system development and circadian rhythm, which contributed to the transition from terrestrial to fully aquatic lifestyles. The African manatee currently possesses a small effective population size and low genome-wide heterozygosity. Overall, our study provides genetic resources for understanding the evolutionary characteristics and conservation efforts of this species.
Collapse
Affiliation(s)
- Xin Huang
- Center for Evolution and Conservation Biology, Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 511458, China
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guixin Dong
- Guangdong Chimelong Group, Co., Ltd., Guangzhou 511400, China
| | - Huizhong Fan
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Wenliang Zhou
- Center for Evolution and Conservation Biology, Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 511458, China
| | - Guangping Huang
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Jiangxi Provincial Key Laboratory of Conservation Biology, College of Forestry, Jiangxi Agricultural University, Nanchang 330045, China
| | - Dengfeng Guan
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Delu Zhang
- Chimelong Ocean Kingdom, Zhuhai 519000, China
| | - Fuwen Wei
- Center for Evolution and Conservation Biology, Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 511458, China
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Jiangxi Provincial Key Laboratory of Conservation Biology, College of Forestry, Jiangxi Agricultural University, Nanchang 330045, China
| |
Collapse
|
3
|
Kang J, Huang G, Ma L, Tong Y, Shahapal A, Chen P, Shen J. Cell-autonomous role of leucine-rich repeat kinase in the protection of dopaminergic neuron survival. eLife 2024; 12:RP92673. [PMID: 38856715 PMCID: PMC11164531 DOI: 10.7554/elife.92673] [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] [Academic Contribution Register] [Indexed: 06/11/2024] Open
Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common genetic cause of Parkinson's disease (PD). However, whether LRRK2 mutations cause PD and degeneration of dopaminergic (DA) neurons via a toxic gain-of-function or a loss-of-function mechanism is unresolved and has pivotal implications for LRRK2-based PD therapies. In this study, we investigate whether Lrrk2 and its functional homolog Lrrk1 play a cell-intrinsic role in DA neuron survival through the development of DA neuron-specific Lrrk conditional double knockout (cDKO) mice. Unlike Lrrk germline DKO mice, DA neuron-restricted Lrrk cDKO mice exhibit normal mortality but develop age-dependent loss of DA neurons, as shown by the progressive reduction of DA neurons in the substantia nigra pars compacta (SNpc) at the ages of 20 and 24 months. Moreover, DA neurodegeneration is accompanied with increases in apoptosis and elevated microgliosis in the SNpc as well as decreases in DA terminals in the striatum, and is preceded by impaired motor coordination. Taken together, these findings provide the unequivocal evidence for the cell-intrinsic requirement of LRRK in DA neurons and raise the possibility that LRRK2 mutations may impair its protection of DA neurons, leading to DA neurodegeneration in PD.
Collapse
Affiliation(s)
- Jongkyun Kang
- Department of Neurology, Brigham and Women’s HospitalBostonUnited States
| | - Guodong Huang
- Department of Neurology, Brigham and Women’s HospitalBostonUnited States
| | - Long Ma
- Department of Neurology, Brigham and Women’s HospitalBostonUnited States
| | - Youren Tong
- Department of Neurology, Brigham and Women’s HospitalBostonUnited States
| | - Anu Shahapal
- Department of Neurology, Brigham and Women’s HospitalBostonUnited States
| | - Phoenix Chen
- Department of Neurology, Brigham and Women’s HospitalBostonUnited States
| | - Jie Shen
- Department of Neurology, Brigham and Women’s HospitalBostonUnited States
- Program in Neuroscience, Harvard Medical SchoolBostonUnited States
| |
Collapse
|
4
|
Kang J, Huang G, Ma L, Tong Y, Shahapal A, Chen P, Shen J. Cell autonomous role of leucine-rich repeat kinase in protection of dopaminergic neuron survival. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.06.561293. [PMID: 37873418 PMCID: PMC10592668 DOI: 10.1101/2023.10.06.561293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 10/25/2023]
Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common genetic cause of Parkinson's disease (PD), which is the leading neurodegenerative movement disorder characterized by the progressive loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc). However, whether LRRK2 mutations cause PD and degeneration of DA neurons via a toxic gain-of-function or a loss-of-function mechanism is unresolved and has pivotal implications for LRRK2-based PD therapies. In this study, we investigate whether LRRK2 and its functional homologue LRRK1 play an essential, intrinsic role in DA neuron survival through the development of DA neuron-specific LRRK conditional double knockout (cDKO) mice. We first generated and characterized floxed LRRK1 and LRRK2 mice and then confirmed that germline deletions of the floxed LRRK1 and LRRK2 alleles result in null mutations, as evidenced by the absence of LRRK1 and LRRK2 mRNA and protein in the respective homozygous deleted mutant mice. We further examined the specificity of Cre-mediated recombination driven by the dopamine transporter-Cre (DAT-Cre) knockin (KI) allele using a GFP reporter line and confirmed that DAT-Cre-mediated recombination is restricted to DA neurons in the SNpc. Crossing these validated floxed LRRK1 and LRRK2 mice with DAT-Cre KI mice, we then generated DA neuron-restricted LRRK cDKO mice and further showed that levels of LRRK1 and LRRK2 are reduced in dissected ventral midbrains of LRRK cDKO mice. While DA neuron-restricted LRRK cDKO mice of both sexes exhibit normal mortality and body weight, they develop age-dependent loss of DA neurons in the SNpc, as demonstrated by the progressive reduction of DA neurons in the SNpc of LRRK cDKO mice at the ages of 20 and 24 months but the unaffected number of DA neurons at the age of 15 months. Moreover, DA neurodegeneration is accompanied with increases of apoptosis and elevated microgliosis in the SNpc as well as decreases of DA terminals in the striatum, and is preceded by impaired motor coordination. Taken together, these findings provide the unequivocal evidence for the importance of LRRK in DA neurons and raise the possibility that LRRK2 mutations may impair its protection of DA neurons, leading to DA neurodegeneration in PD.
Collapse
Affiliation(s)
- Jongkyun Kang
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, United States of America
| | - Guodong Huang
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, United States of America
| | - Long Ma
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, United States of America
| | - Youren Tong
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, United States of America
| | - Anu Shahapal
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, United States of America
| | - Phoenix Chen
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, United States of America
| | - Jie Shen
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, United States of America
- Program in Neuroscience, Harvard Medical School, Boston, MA 02115, United States of America
| |
Collapse
|
5
|
Xing W, Pourteymoor S, Udayakumar A, Chen Y, Mohan S. Targeted Overexpression of Claudin 11 in Osteoblasts Increases Trabecular Bone Mass by Stimulating Osteogenesis at the Expense of Adipogenesis in Mice. BIOLOGY 2024; 13:108. [PMID: 38392326 PMCID: PMC10886834 DOI: 10.3390/biology13020108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 12/08/2023] [Revised: 02/06/2024] [Accepted: 02/07/2024] [Indexed: 02/24/2024]
Abstract
Mice lacking Claudin11 (Cldn11) manifest reduced trabecular bone mass. However, the impact of Cldn11 expression in osteoblasts in vivo remains understudied. Herein, we generated osteoblast-specific transgenic (Tg) mice expressing Cldn11 and characterized their skeletal phenotype. Micro-CT analyses of the distal metaphysis of the femur showed a 50% and a 38% increase in trabecular bone mass in Tg male and female mice, respectively, due to a significant increase in trabecular number and a reduction in trabecular separation. Histomorphometry and serum biomarker studies uncovered that increased trabecular bone mass in Cldn11 Tg mice was the consequence of enhanced bone formation. Accordingly, an abundance of bone formation (Alp, Bsp), but not bone resorption (Ctsk), markers were augmented in the femurs of Cldn11 Tg mice. Since the trabecular bone density is known to inversely correlate with the amount of marrow adipose tissue (MAT), we measured the MAT in osmium-tetroxide-labeled bones by micro-CT scanning. We found 86% less MAT in the proximal tibia of the Tg males. Consistently, the expression levels of the adipogenic markers, adiponectin and leptin, were 50% lower in the femurs of the Tg males. Our data are consistent with the possibility that claudin11 exerts anabolic effects in osteoblastic lineage cells that act via promoting the differentiation of marrow stem cells towards osteoblasts at the expense of adipocytes.
Collapse
Affiliation(s)
- Weirong Xing
- Musculoskeletal Disease Center, Loma Linda VA Healthcare System, Loma Linda, CA 92357, USA
- Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Sheila Pourteymoor
- Musculoskeletal Disease Center, Loma Linda VA Healthcare System, Loma Linda, CA 92357, USA
| | - Anakha Udayakumar
- Musculoskeletal Disease Center, Loma Linda VA Healthcare System, Loma Linda, CA 92357, USA
- Graduate School, Loma Linda University, Loma Linda, CA 92354, USA
| | - Yian Chen
- Musculoskeletal Disease Center, Loma Linda VA Healthcare System, Loma Linda, CA 92357, USA
| | - Subburaman Mohan
- Musculoskeletal Disease Center, Loma Linda VA Healthcare System, Loma Linda, CA 92357, USA
- Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| |
Collapse
|
6
|
Pieridou C, Sabir A, Lancashire J, Liang Y, McMillan K, Shaw N, Uday S. Case Report: Osteosclerotic metaphyseal dysplasia with optic nerve involvement and progressive osteonecrosis of the jaw due to a novel LRRK1 mutation. Front Endocrinol (Lausanne) 2023; 14:1258340. [PMID: 37920250 PMCID: PMC10619726 DOI: 10.3389/fendo.2023.1258340] [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] [Academic Contribution Register] [Received: 07/13/2023] [Accepted: 09/18/2023] [Indexed: 11/04/2023] Open
Abstract
Background Osteosclerotic metaphyseal dysplasia (OSMD, OMIM 615198) is an extremely rare autosomal recessive osteopetrosis disorder resulting in a distinctive pattern of osteosclerosis of the metaphyseal margins of long tubular bones. To date, only thirteen cases have been reported (eight molecularly confirmed). Five homozygous sequence variants in the leucine-rich repeat kinase 1 (LRRK1) gene have been identified to cause OSMD. We present two male siblings with OSMD with a novel LRRK1 variant. Cases The index case, now aged 6 years, was referred aged 9 months when diffuse sclerosis of the ribs and vertebral bodies, suggestive of osteopetrosis, was incidentally identified on a chest radiograph for suspected lower respiratory tract infection. Parents were consanguineous and of Pakistani origin. Further evaluation revealed developmental delay, nystagmus with bilateral optic nerve hypoplasia and severe visual impairment. Skeletal survey confirmed typical changes of OSMD, with widespread diffuse sclerosis and Erlenmeyer flask deformity of long bones. His older sibling, now aged 12 years, was 7 years at the time of referral and had similar clinical course and skeletal findings. Additionally, he had a chronic progressive osteonecrosis of the left mandible that required debridement, debulking and long-term antibiotics. Skeletal survey revealed findings similar to his sibling. Neither sibling had significant skeletal fractures or seizures. Unlike most previous reports suggesting sparing of the skull and lack of visual impairment, our patients had evidence of osteosclerosis of the cranium. Genetic screening for the common autosomal recessive and dominant pathogenic variants of osteopetrosis was negative. Whole Exome Sequencing (WES) followed by Sanger sequencing, identified a novel homozygous LRRK1 c.2506C>T p. (Gln836Ter) nonsense variant predicted to result in premature truncation of LRRK1 transcript. Conclusion Our cases confirm the autosomal recessive inheritance and expand the spectrum of genotype and phenotype of OSMD reported in the literature. Increasing reports of LRRK1 variants in this phenotype raise the question of whether LRRK1 should be included in targeted osteopetrosis panels. Bone histology in previous cases has shown this to be an osteoclast rich form of osteopetrosis raising the possibility that haematopoietic stem cell transplantation may be an appropriate treatment modality.
Collapse
Affiliation(s)
- Chariklia Pieridou
- Department of Endocrinology and Diabetes, Birmingham Women’s and Children’s Hospital, Birmingham, United Kingdom
| | - Ataf Sabir
- Department of Clinical Genetics, Birmingham Women’s and Children’s Hospital, Birmingham, United Kingdom
| | - Jonathan Lancashire
- Department of Hematology, Birmingham Women’s and Children’s Hospital, Birmingham, United Kingdom
| | - Yifan Liang
- Department of Paediatric Palliative Medicine, Birmingham Women’s and Children’s and Community Healthcare Trusts, Birmingham, United Kingdom
| | - Kevin McMillan
- Department of Oral and Maxillofacial Surgery, Birmingham Women’s and Children’s Hospital, Birmingham, United Kingdom
| | - Nick Shaw
- Department of Endocrinology and Diabetes, Birmingham Women’s and Children’s Hospital, Birmingham, United Kingdom
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
| | - Suma Uday
- Department of Endocrinology and Diabetes, Birmingham Women’s and Children’s Hospital, Birmingham, United Kingdom
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
7
|
Sivakumaran TA, Grebe TA. 15q26.3 deletions distal to IGF1R cause growth retardation, congenital heart defect and skeletal anomalies: Case report and review of literature. Am J Med Genet A 2023; 191:2392-2397. [PMID: 37434556 DOI: 10.1002/ajmg.a.63350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/10/2023] [Revised: 06/23/2023] [Accepted: 06/28/2023] [Indexed: 07/13/2023]
Abstract
15q26 deletion is a rare genomic disorder characterized by intrauterine and postnatal growth retardation, microcephaly, intellectual disability, and congenital malformations. Here, we report a 4-month-old female with intrauterine growth retardation, short stature, pulmonary hypertension, atrial septal defect and congenital bowing of long bones of the legs. Chromosomal microarray analysis showed a de novo deletion of approximately 2.1 Mb at 15q26.3 region that does not include IGF1R. Our analysis of patients documented in the literature and the DECIPHER database with 15q26 deletions distal to IGF1R, including 10 patients with de novo pure deletions, allowed us to define the smallest region of overlap to 686 kb. This region includes ALDH1A3, LRRK1, CHSY1, SELENOS, SNRPA1, and PCSK6. We propose haploinsufficiency of one or more genes, besides IGF1R, within this region may contribute to the clinical findings in patients with 15q26.3 deletion.
Collapse
Affiliation(s)
- Theru A Sivakumaran
- Division of Pathology and Laboratory Medicine, Phoenix Children's Hospital, Phoenix, Arizona, USA
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, USA
- Department of Pathology, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, USA
- Department of Pathology, Creighton University School of Medicine, Phoenix, Arizona, USA
| | - Theresa A Grebe
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, USA
- Division of Genetics and Metabolism, Phoenix Children's Hospital, Phoenix, Arizona, USA
| |
Collapse
|
8
|
Metcalfe RD, Martinez Fiesco JA, Bonet-Ponce L, Kluss JH, Cookson MR, Zhang P. Structure and regulation of full-length human leucine-rich repeat kinase 1. Nat Commun 2023; 14:4797. [PMID: 37558661 PMCID: PMC10412621 DOI: 10.1038/s41467-023-40532-2] [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] [Academic Contribution Register] [Received: 12/13/2022] [Accepted: 07/26/2023] [Indexed: 08/11/2023] Open
Abstract
The human leucine-rich repeat kinases (LRRKs), LRRK1 and LRRK2 are large and unusually complex multi-domain kinases, which regulate fundamental cellular processes and have been implicated in human disease. Structures of LRRK2 have recently been determined, but the structure and molecular mechanisms regulating the activity of the LRRK1 as well as differences in the regulation of LRRK1 and LRRK2 remain unclear. Here, we report a cryo-EM structure of the LRRK1 monomer and a lower-resolution cryo-EM map of the LRRK1 dimer. The monomer structure, in which the kinase is in an inactive conformation, reveals key interdomain interfaces that control kinase activity as we validate experimentally. Both the LRRK1 monomer and dimer are structurally distinct compared to LRRK2. Overall, our results provide structural insights into the activation of the human LRRKs, which advance our understanding of their physiological and pathological roles.
Collapse
Affiliation(s)
- Riley D Metcalfe
- Center for Structural Biology, Center for Cancer Research, National Cancer Institute, Frederick, MD, 21702, USA
| | - Juliana A Martinez Fiesco
- Center for Structural Biology, Center for Cancer Research, National Cancer Institute, Frederick, MD, 21702, USA
| | - Luis Bonet-Ponce
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jillian H Kluss
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Mark R Cookson
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ping Zhang
- Center for Structural Biology, Center for Cancer Research, National Cancer Institute, Frederick, MD, 21702, USA.
| |
Collapse
|
9
|
Tang L, Li B, Su Q, Chen X, He R. Identification of hub genes and therapeutic drugs in osteonecrosis of the femoral head through integrated bioinformatics analysis and literature mining. Sci Rep 2023; 13:11972. [PMID: 37488209 PMCID: PMC10366127 DOI: 10.1038/s41598-023-39258-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/27/2023] [Accepted: 07/22/2023] [Indexed: 07/26/2023] Open
Abstract
Osteonecrosis of the femoral head (ONFH) is a multifactorial disease leading to severely limited function. By far, the etiology and pathogenesis of ONFH are not fully understood, and surgery is the only effective way to treat ONFH. This study aims to identify hub genes and therapeutic drugs in ONFH. Two gene expression profiles were downloaded from the gene expression omnibus database, and the hub genes and candidate drugs for ONFH were identified through integrated bioinformatics analysis and cross-validated by literature mining. A total of 159 DEGs were identified. PTGS2, LRRK2, ANXA5, IGF1R, MCL1, TIMP2, LYN, CD68, CBL, and RUNX2 were validated as 10 hub genes, which has considerable implications for future genetic research and related research fields of ONFH. Our findings indicate that 85 drugs interact with ONFH, with most drugs exhibiting a positive impact on ONFH by promoting osteogenesis and angiogenesis or inhibiting microcirculation embolism, rather than being anti-inflammatory. Our study provides novel insights into the pathogenesis, prevention, and treatment of ONFH.
Collapse
Affiliation(s)
- Lan Tang
- Department of Orthopedic, The Second Affiliated Hospital, Zhejiang University School of Medicine, #88 Jiefang Road, Hangzhou City, 310001, Zhejiang Province, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China
| | - Bin Li
- Department of Orthopedic, The Second Affiliated Hospital, Zhejiang University School of Medicine, #88 Jiefang Road, Hangzhou City, 310001, Zhejiang Province, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China
| | - Qiuming Su
- Department of Hepatopancreatobiliary Surgery, The First People's Hospital of Kunming, Calmette Hospital, Kunming City, Yunnan Province, China
| | - Xi Chen
- Department of Orthopedic, The Second Affiliated Hospital, Zhejiang University School of Medicine, #88 Jiefang Road, Hangzhou City, 310001, Zhejiang Province, People's Republic of China
- Department of Epidemiology and Statistics, School of Public Health, Medical College, Zhejiang University, Hangzhou City, Zhejiang Province, China
| | - Rongxin He
- Department of Orthopedic, The Second Affiliated Hospital, Zhejiang University School of Medicine, #88 Jiefang Road, Hangzhou City, 310001, Zhejiang Province, People's Republic of China.
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China.
| |
Collapse
|
10
|
Shen S, Si M, Zeng C, Liu EK, Chen Y, Vacher J, Zhao H, Mohan S, Xing W. Leucine Repeat Rich Kinase 1 Controls Osteoclast Activity by Managing Lysosomal Trafficking and Secretion. BIOLOGY 2023; 12:511. [PMID: 37106712 PMCID: PMC10135754 DOI: 10.3390/biology12040511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 02/28/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 03/31/2023]
Abstract
We previously demonstrated that mice with targeted deletion of the leucine repeat rich kinase 1 (Lrrk1) gene were osteopetrotic due to the failure of osteoclasts to resorb bone. To determine how LRRK1 regulates osteoclast activity, we examined the intracellular and extracellular acidification with an acidotropic probe, acridine orange, in live osteoclasts on bone slices. We examined lysosome distribution in osteoclasts by localization of LAMP-2, cathepsin K, and v-ATPase by immunofluorescent staining with specific antibodies. We found that both vertical and horizontal cross-sectional images of the wild-type (WT) osteoclasts showed orange-staining of the intracellular acidic vacuoles/lysosomes dispersed to the ruffled border. By contrast, the LRRK1 deficient osteoclasts exhibited fluorescent orange staining in the cytoplasm away from the extracellular lacunae because of an altered distribution of the acidic vacuoles/lysosomes. In addition, WT osteoclasts displayed a peripheral distribution of LAMP-2 positive lysosomes with a typical actin ring. The clustered F-actin constitutes a peripheral sealing zone and a ruffled border which was stretched out into a resorption pit. The LAMP-2 positive lysosomes were also distributed to the sealing zone, and the cell was associated with a resorption pit. By contrast, LRRK1-deficient osteoclasts showed diffused F-actin throughout the cytoplasm. The sealing zone was weak and not associated with a resorption pit. LAMP-2 positive lysosomes were also diffuse in the cytoplasm and were not distributed to the ruffled border. Although the LRRK1-deficient osteoclast expressed normal levels of cathepsin K and v-ATPase, the lysosomal-associated cathepsin K and v-ATPase were not accumulated at the ruffled border in Lrrk1 KO osteoclasts. Our data indicate that LRRK1 controls osteoclast activity by regulating lysosomal distribution, acid secretion, and protease exocytosis.
Collapse
Affiliation(s)
- Sandi Shen
- Musculoskeletal Disease Center, Jerry L Pettis VA Medical Center, Loma Linda, CA 92357, USA
- Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Mingjue Si
- Musculoskeletal Disease Center, Jerry L Pettis VA Medical Center, Loma Linda, CA 92357, USA
- Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Canjun Zeng
- Musculoskeletal Disease Center, Jerry L Pettis VA Medical Center, Loma Linda, CA 92357, USA
- Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Elaine K. Liu
- Musculoskeletal Disease Center, Jerry L Pettis VA Medical Center, Loma Linda, CA 92357, USA
| | - Yian Chen
- Musculoskeletal Disease Center, Jerry L Pettis VA Medical Center, Loma Linda, CA 92357, USA
| | - Jean Vacher
- Institut de Recherches Cliniques de Montreal, Montreal, QC H2W 1R7, Canada
- Département de Médecine, Université de Montréal, Montréal, QC H2W 1R7, Canada
| | - Haibo Zhao
- Southern California Institute for Research and Education, Long Beach, CA 90822, USA
| | - Subburaman Mohan
- Musculoskeletal Disease Center, Jerry L Pettis VA Medical Center, Loma Linda, CA 92357, USA
- Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Weirong Xing
- Musculoskeletal Disease Center, Jerry L Pettis VA Medical Center, Loma Linda, CA 92357, USA
- Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| |
Collapse
|
11
|
Brommage R, Liu J, Powell DR. Skeletal phenotypes in secreted frizzled-related protein 4 gene knockout mice mimic skeletal architectural abnormalities in subjects with Pyle's disease from SFRP4 mutations. Bone Res 2023; 11:9. [PMID: 36808149 PMCID: PMC9941579 DOI: 10.1038/s41413-022-00242-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/29/2022] [Revised: 09/26/2022] [Accepted: 11/03/2022] [Indexed: 02/22/2023] Open
Abstract
Mutations in SFRP4 cause Pyle's bone disease with wide metaphyses and increased skeletal fragility. The WNT signaling pathway plays important roles in determining skeletal architecture and SFRP4 is a secreted Frizzled decoy receptor that inhibits WNT signaling. Seven cohorts of male and female Sfrp4 gene knockout mice, examined through 2 years of age, had a normal lifespan but showed cortical and trabecular bone phenotypes. Mimicking human Erlenmeyer flask deformities, bone cross-sectional areas were elevated 2-fold in the distal femur and proximal tibia but only 30% in femur and tibia shafts. Reduced cortical bone thickness was observed in the vertebral body, midshaft femur and distal tibia. Elevated trabecular bone mass and numbers were observed in the vertebral body, distal femur metaphysis and proximal tibia metaphysis. Midshaft femurs retained extensive trabecular bone through 2 years of age. Vertebral bodies had increased compressive strength, but femur shafts had reduced bending strength. Trabecular, but not cortical, bone parameters in heterozygous Sfrp4 mice were modestly affected. Ovariectomy resulted in similar declines in both cortical and trabecular bone mass in wild-type and Sfrp4 KO mice. SFRP4 is critical for metaphyseal bone modeling involved in determining bone width. Sfrp4 KO mice show similar skeletal architecture and bone fragility deficits observed in patients with Pyle's disease with SFRP4 mutations.
Collapse
Affiliation(s)
- Robert Brommage
- Department of Metabolism Research, Lexicon Pharmaceuticals, The Woodlands, TX, 77381, USA.
- BoneGenomics, The Woodlands, TX, USA.
| | - Jeff Liu
- Department of Metabolism Research, Lexicon Pharmaceuticals, The Woodlands, TX, 77381, USA.
- Biogen, Cambridge, MA, USA.
| | - David R Powell
- Department of Metabolism Research, Lexicon Pharmaceuticals, The Woodlands, TX, 77381, USA.
| |
Collapse
|
12
|
Turan S. Osteopetrosis: Gene-based nosology and significance Dysosteosclerosis. Bone 2023; 167:116615. [PMID: 36402365 DOI: 10.1016/j.bone.2022.116615] [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] [Academic Contribution Register] [Received: 07/03/2022] [Revised: 11/09/2022] [Accepted: 11/12/2022] [Indexed: 11/18/2022]
Abstract
Dysosteosclerosis (DSS) refers to skeletal dysplasias that radiographically feature focal appendicular osteosclerosis with variable platyspondyly. Genetic heterogeneity is increasingly reported for the DSS phenotype and now involves mutations of SLC29A3, TNFRSF11A, TCIRG1, LRRK1, and CSF1R. Typical radiological findings are widened radiolucent long bones with thin cortices yet dense irregular metaphyses, flattened vertebral bodies, dense ribs, and multiple fractures. However, the radiographic features of DSS evolve, and the metaphyseal and/or appendicular osteosclerosis variably fades with increasing patient age, likely due to some residual osteoclast function. Fractures are the principal presentation of DSS, and may even occur in infancy with SLC29A3-associated DSS. Cranial base sclerosis can lead to cranial nerve palsies such as optic atrophy, and may be the initial presentation, though not observed with SLC29A3-associated DSS. Gene-specific extra-skeletal features can be the main complication in some forms of DSS such as CSF1R- associated DSS. Further genetic heterogeneity is likely, especially for X-linked recessive DSS and cases currently with an unknown genetic defect. Distinguishing DSS can be challenging due to variable clinical and radiological features and an evolving phenotype. However, defining the DSS phenotype is important for predicting complications, prognosis, and instituting appropriate health surveillance and treatment.
Collapse
Affiliation(s)
- Serap Turan
- Pediatric Endocrinology and Diabetes, Marmara University School of Medicine, Istanbul, Turkey.
| |
Collapse
|
13
|
Xing W, Larkin D, Pourteymoor S, Tambunan W, Gomez GA, Liu EK, Mohan S. Lack of Skeletal Effects in Mice with Targeted Disruptionof Prolyl Hydroxylase Domain 1 ( Phd1) Gene Expressed in Chondrocytes. Life (Basel) 2022; 13:106. [PMID: 36676055 PMCID: PMC9862499 DOI: 10.3390/life13010106] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/30/2022] [Revised: 12/16/2022] [Accepted: 12/28/2022] [Indexed: 01/01/2023] Open
Abstract
The critical importance of hypoxia-inducible factor (HIF)s in the regulation of endochondral bone formation is now well established. HIF protein levels are closely regulated by the prolyl hydroxylase domain-containing protein (PHD) mediated ubiquitin-proteasomal degradation pathway. Of the three PHD family members expressed in bone, we previously showed that mice with conditional disruption of the Phd2 gene in chondrocytes led to a massive increase in the trabecular bone mass of the long bones. By contrast, loss of Phd3 expression in chondrocytes had no skeletal effects. To investigate the role of Phd1 expressed in chondrocytes on skeletal development, we conditionally disrupted the Phd1 gene in chondrocytes by crossing Phd1 floxed mice with Collagen 2α1-Cre mice for evaluation of a skeletal phenotype. At 12 weeks of age, neither body weight nor body length was significantly different in the Cre+; Phd1flox/flox conditional knockout (cKO) mice compared to Cre−; Phd1flox/flox wild-type (WT) control mice. Micro-CT measurements revealed significant gender differences in the trabecular bone volume adjusted for tissue volume at the secondary spongiosa of the femur and the tibia for both genotypes, but no genotype differences were found for any of the trabecular bone measurements of either femur or tibia. Similarly, cortical bone parameters were not affected in the Phd1 cKO mice compared to control mice. Histomorphometric analyses revealed no significant differences in bone area, bone formation rate or mineral apposition rate in the secondary spongiosa of femurs between cKO and WT control mice. Loss of Phd1 expression in chondrocytes did not affect the expression of markers of chondrocytes (collage 2, collagen 10) or osteoblasts (alkaline phosphatase, bone sialoprotein) in the bones of cKO mice. Based on these and our published data, we conclude that of the three PHD family members, only Phd2 expressed in chondrocytes regulates endochondral bone formation and development of peak bone mass in mice.
Collapse
Affiliation(s)
- Weirong Xing
- Musculoskeletal Disease Center, Loma Linda VA Healthcare System, Loma Linda, CA 92357, USA
- Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Destiney Larkin
- Musculoskeletal Disease Center, Loma Linda VA Healthcare System, Loma Linda, CA 92357, USA
| | - Sheila Pourteymoor
- Musculoskeletal Disease Center, Loma Linda VA Healthcare System, Loma Linda, CA 92357, USA
| | - William Tambunan
- Musculoskeletal Disease Center, Loma Linda VA Healthcare System, Loma Linda, CA 92357, USA
| | - Gustavo A. Gomez
- Musculoskeletal Disease Center, Loma Linda VA Healthcare System, Loma Linda, CA 92357, USA
| | - Elaine K. Liu
- Musculoskeletal Disease Center, Loma Linda VA Healthcare System, Loma Linda, CA 92357, USA
| | - Subburaman Mohan
- Musculoskeletal Disease Center, Loma Linda VA Healthcare System, Loma Linda, CA 92357, USA
- Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| |
Collapse
|
14
|
Gomez GA, Rundle CH, Xing W, Kesavan C, Pourteymoor S, Lewis RE, Powell DR, Mohan S. Contrasting effects of <i>Ksr2</i>, an obesity gene, on trabecular bone volume and bone marrow adiposity. eLife 2022; 11:82810. [PMID: 36342465 PMCID: PMC9640193 DOI: 10.7554/elife.82810] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/18/2022] [Accepted: 10/06/2022] [Indexed: 11/25/2022] Open
Abstract
Pathological obesity and its complications are associated with an increased propensity for bone fractures. Humans with certain genetic polymorphisms at the kinase suppressor of ras2 (KSR2) locus develop severe early-onset obesity and type 2 diabetes. Both conditions are phenocopied in mice with <i>Ksr2</i> deleted, but whether this affects bone health remains unknown. Here we studied the bones of global <i>Ksr2</i> null mice and found that <i>Ksr2</i> negatively regulates femoral, but not vertebral, bone mass in two genetic backgrounds, while the paralogous gene, <i>Ksr1</i>, was dispensable for bone homeostasis. Mechanistically, KSR2 regulates bone formation by influencing adipocyte differentiation at the expense of osteoblasts in the bone marrow. Compared with <i>Ksr2</i>'s known role as a regulator of feeding by its function in the hypothalamus, pair-feeding and osteoblast-specific conditional deletion of <i>Ksr2</i> reveals that <i>Ksr2</i> can regulate bone formation autonomously. Despite the gains in appendicular bone mass observed in the absence of <i>Ksr2</i>, bone strength, as well as fracture healing response, remains compromised in these mice. This study highlights the interrelationship between adiposity and bone health and provides mechanistic insights into how <i>Ksr2</i>, an adiposity and diabetic gene, regulates bone metabolism.
Collapse
Affiliation(s)
| | - Charles H Rundle
- VA Loma Linda Healthcare SystemLoma LindaUnited States,Loma Linda University Medical CenterLoma LindaUnited States
| | - Weirong Xing
- VA Loma Linda Healthcare SystemLoma LindaUnited States,Loma Linda University Medical CenterLoma LindaUnited States
| | - Chandrasekhar Kesavan
- VA Loma Linda Healthcare SystemLoma LindaUnited States,Loma Linda University Medical CenterLoma LindaUnited States
| | | | | | | | - Subburaman Mohan
- VA Loma Linda Healthcare SystemLoma LindaUnited States,Loma Linda University Medical CenterLoma LindaUnited States
| |
Collapse
|
15
|
Chorin O, Chowers G, Agbariah R, Karklinsky S, Barel O, Bar-Joseph I, Reznik-Wolf H, Shamash J, Pode-Shakked B, Jacobson JM, Huna-Baron R, Redler Y, Tirosh I, Vivante A, Raas-Rothschild A. Broadening the phenotype of LRRK1 mutations - Features of malignant osteopetrosis and optic nerve atrophy with intrafamilial variable expressivity. Eur J Med Genet 2021; 65:104383. [PMID: 34798323 DOI: 10.1016/j.ejmg.2021.104383] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/26/2021] [Revised: 08/26/2021] [Accepted: 11/07/2021] [Indexed: 11/03/2022]
Abstract
Osteosclerotic metaphyseal dysplasia is a rare disorder which features osteosclerosis involving long bones, vertebrae, ribs, clavicles and the iliac crests. Additional features which have variably been reported include developmental delay, short stature, hypotonia and seizures. The disease is caused by pathogenic variants in the LRRK1 gene, and inherited in an autosomal recessive manner. We report three siblings (ages 14 years, 11.5 years and 0.9 years), born to consanguineous parents of Arab-Muslim descent, harboring a homozygous pathogenic variant in the LRRK1 gene (Chr15:101068759 AGGGGCT>A, c.5965_5970del TGGGGC, p.Trp1989Gly1990del). The patients displayed variable degrees of skeletal dysplasia, with the oldest sibling most severely affected, and the youngest infant with minor skeletal involvement. Two of the siblings exhibited normal neurological development, while the youngest sibling exhibited global developmental delay. None of the siblings had seizures; however, two of them exhibited nystagmus. Optic nerve involvement has not previously been reported to be part of the clinical spectrum of this disease. The degree of optic nerve involvement did not correlate with the degree of skeletal involvement. This indicates both intra-familial variable expressivity along with a broadening of the spectrum of LRRK1-associated disease. These findings warrant reconsideration of therapeutic strategies, including the possibility of hematopoietic stem cell transplantation (HSCT) as is performed in cases of malignant and intermediate forms of osteopetrosis.
Collapse
Affiliation(s)
- Odelia Chorin
- Institute of Rare Diseases, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Israel; The Genomic Unit, Sheba Cancer Research Center, Sheba Medical Center, Tel-Hashomer, Israel
| | - Guy Chowers
- Department of Pediatrics B, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Israel
| | - Rawan Agbariah
- Department of Pediatrics B, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Israel
| | - Shani Karklinsky
- Department of Pediatrics B, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Israel
| | - Ortal Barel
- The Genomic Unit, Sheba Cancer Research Center, Sheba Medical Center, Tel-Hashomer, Israel; The Wohl Institute of Translational Medicine, Sheba Medical Center, Tel-Hashomer, Israel
| | - Ifat Bar-Joseph
- The Genomic Unit, Sheba Cancer Research Center, Sheba Medical Center, Tel-Hashomer, Israel; The Wohl Institute of Translational Medicine, Sheba Medical Center, Tel-Hashomer, Israel
| | - Haike Reznik-Wolf
- The Danek Gertner Institute of Human Genetics, Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Jana Shamash
- The Danek Gertner Institute of Human Genetics, Sheba Medical Center, Tel-Hashomer, Israel
| | - Ben Pode-Shakked
- Institute of Rare Diseases, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Israel; Department of Pediatrics B, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Israel; Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Jeffrey M Jacobson
- Pediatric Imaging Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Ruth Huna-Baron
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Neuro-Ophthalmology Unit, Sheba Medical Center, Tel-Hashomer, Israel
| | - Yael Redler
- Neuro-Ophthalmology Unit, Sheba Medical Center, Tel-Hashomer, Israel
| | - Irit Tirosh
- Department of Pediatrics B, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Asaf Vivante
- Department of Pediatrics B, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Israel; Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel; Pediatric Nephrology Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Annick Raas-Rothschild
- Institute of Rare Diseases, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| |
Collapse
|
16
|
Suzzi S, Ahrendt R, Hans S, Semenova SA, Chekuru A, Wirsching P, Kroehne V, Bilican S, Sayed S, Winkler S, Spieß S, Machate A, Kaslin J, Panula P, Brand M. Deletion of lrrk2 causes early developmental abnormalities and age-dependent increase of monoamine catabolism in the zebrafish brain. PLoS Genet 2021; 17:e1009794. [PMID: 34516550 PMCID: PMC8459977 DOI: 10.1371/journal.pgen.1009794] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/14/2021] [Revised: 09/23/2021] [Accepted: 08/24/2021] [Indexed: 12/12/2022] Open
Abstract
LRRK2 gain-of-function is considered a major cause of Parkinson's disease (PD) in humans. However, pathogenicity of LRRK2 loss-of-function in animal models is controversial. Here we show that deletion of the entire zebrafish lrrk2 locus elicits a pleomorphic transient brain phenotype in maternal-zygotic mutant embryos (mzLrrk2). In contrast to lrrk2, the paralog gene lrrk1 is virtually not expressed in the brain of both wild-type and mzLrrk2 fish at different developmental stages. Notably, we found reduced catecholaminergic neurons, the main target of PD, in specific cell populations in the brains of mzLrrk2 larvae, but not adult fish. Strikingly, age-dependent accumulation of monoamine oxidase (MAO)-dependent catabolic signatures within mzLrrk2 brains revealed a previously undescribed interaction between LRRK2 and MAO biological activities. Our results highlight mzLrrk2 zebrafish as a tractable tool to study LRRK2 loss-of-function in vivo, and suggest a link between LRRK2 and MAO, potentially of relevance in the prodromic stages of PD.
Collapse
Affiliation(s)
- Stefano Suzzi
- Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Technische Universität (TU) Dresden, Dresden, Germany
| | - Reiner Ahrendt
- Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Technische Universität (TU) Dresden, Dresden, Germany
| | - Stefan Hans
- Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Technische Universität (TU) Dresden, Dresden, Germany
| | - Svetlana A. Semenova
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Avinash Chekuru
- Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Technische Universität (TU) Dresden, Dresden, Germany
| | - Paul Wirsching
- Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Technische Universität (TU) Dresden, Dresden, Germany
| | - Volker Kroehne
- Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Technische Universität (TU) Dresden, Dresden, Germany
| | - Saygın Bilican
- Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Technische Universität (TU) Dresden, Dresden, Germany
| | - Shady Sayed
- Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Technische Universität (TU) Dresden, Dresden, Germany
| | - Sylke Winkler
- Max Planck Institute of Molecular Cell Biology and Genetics (MPI-CBG), Dresden, Germany
| | - Sandra Spieß
- Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Technische Universität (TU) Dresden, Dresden, Germany
| | - Anja Machate
- Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Technische Universität (TU) Dresden, Dresden, Germany
| | - Jan Kaslin
- Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Technische Universität (TU) Dresden, Dresden, Germany
| | - Pertti Panula
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Michael Brand
- Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Technische Universität (TU) Dresden, Dresden, Germany
- * E-mail:
| |
Collapse
|
17
|
Prolyl Hydroxylase Domain-Containing Protein 3 Gene Expression in Chondrocytes Is Not Essential for Bone Development in Mice. Cells 2021; 10:cells10092200. [PMID: 34571849 PMCID: PMC8470734 DOI: 10.3390/cells10092200] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/02/2021] [Revised: 08/16/2021] [Accepted: 08/20/2021] [Indexed: 01/28/2023] Open
Abstract
We previously showed that conditional disruption of the Phd2 gene in chondrocytes led to a massive increase in long bone trabecular bone mass. Loss of Phd2 gene expression or inhibition of PHD2 activity by a specific inhibitor resulted in a several-fold compensatory increase in Phd3 expression in chondrocytes. To determine if expression of PHD3 plays a role in endochondral bone formation, we conditionally disrupted the Phd3 gene in chondrocytes by crossing Phd3 floxed (Phd3flox/flox) mice with Col2α1-Cre mice. Loss of Phd3 expression in the chondrocytes of Cre+; Phd3flox/flox conditional knockout (cKO) mice was confirmed by real time PCR. At 16 weeks of age, neither body weight nor body length was significantly different in the Phd3 cKO mice compared to Cre−; Phd3flox/flox wild-type (WT) mice. Areal BMD measurements of total body as well as femur, tibia, and lumbar skeletal sites were not significantly different between the cKO and WT mice at 16 weeks of age. Micro-CT measurements revealed significant gender differences in the trabecular bone volume adjusted for tissue volume at the secondary spongiosa of the femur and the tibia for both genotypes, but no genotype difference was found for any of the trabecular bone measurements of either the femur or the tibia. Trabecular bone volume of distal femur epiphysis was not different between cKO and WT mice. Histology analyses revealed Phd3 cKO mice exhibited a comparable chondrocyte differentiation and proliferation, as evidenced by no changes in cartilage thickness and area in the cKO mice as compared to WT littermates. Consistent with the in vivo data, lentiviral shRNA-mediated knockdown of Phd3 expression in chondrocytes did not affect the expression of markers of chondrocyte differentiation (Col2, Col10, Acan, Sox9). Our study found that Phd2 but not Phd3 expressed in chondrocytes regulates endochondral bone formation, and the compensatory increase in Phd3 expression in the chondrocytes of Phd2 cKO mice is not the cause for increased trabecular bone mass in Phd2 cKO mice.
Collapse
|
18
|
Rios JJ, Denton K, Russell J, Kozlitina J, Ferreira CR, Lewanda AF, Mayfield JE, Moresco E, Ludwig S, Tang M, Li X, Lyon S, Khanshour A, Paria N, Khalid A, Li Y, Xie X, Feng JQ, Xu Q, Lu Y, Hammer RE, Wise CA, Beutler B. Germline Saturation Mutagenesis Induces Skeletal Phenotypes in Mice. J Bone Miner Res 2021; 36:1548-1565. [PMID: 33905568 PMCID: PMC8862308 DOI: 10.1002/jbmr.4323] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 10/06/2020] [Revised: 04/07/2021] [Accepted: 04/21/2021] [Indexed: 12/28/2022]
Abstract
Proper embryonic and postnatal skeletal development require coordination of myriad complex molecular mechanisms. Disruption of these processes, through genetic mutation, contributes to variation in skeletal development. We developed a high-throughput N-ethyl-N-nitrosourea (ENU)-induced saturation mutagenesis skeletal screening approach in mice to identify genes required for proper skeletal development. Here, we report initial results from live-animal X-ray and dual-energy X-ray absorptiometry (DXA) imaging of 27,607 G3 mice from 806 pedigrees, testing the effects of 32,198 coding/splicing mutations in 13,020 genes. A total of 39.7% of all autosomal genes were severely damaged or destroyed by mutations tested twice or more in the homozygous state. Results from our study demonstrate the feasibility of in vivo mutagenesis to identify mouse models of skeletal disease. Furthermore, our study demonstrates how ENU mutagenesis provides opportunities to create and characterize putative hypomorphic mutations in developmentally essential genes. Finally, we present a viable mouse model and case report of recessive skeletal disease caused by mutations in FAM20B. Results from this study, including engineered mouse models, are made publicly available via the online Mutagenetix database. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Jonathan J Rios
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, USA.,Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, USA.,McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX, USA.,Department of Orthopaedic Surgery, UT Southwestern Medical Center, Dallas, TX, USA.,Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA
| | - Kristin Denton
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, USA
| | - Jamie Russell
- Center for Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX, USA
| | - Julia Kozlitina
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX, USA
| | - Carlos R Ferreira
- Skeletal Genomics Unit, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Amy F Lewanda
- Rare Disease Institute, Children's National Hospital, Washington, DC, USA
| | - Joshua E Mayfield
- Department of Pharmacology, University of California, San Diego, CA, USA
| | - Eva Moresco
- Center for Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX, USA
| | - Sara Ludwig
- Center for Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX, USA
| | - Miao Tang
- Center for Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX, USA
| | - Xiaohong Li
- Center for Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX, USA
| | - Stephen Lyon
- Center for Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX, USA
| | - Anas Khanshour
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, USA
| | - Nandina Paria
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, USA
| | - Aysha Khalid
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, USA
| | - Yang Li
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, USA
| | - Xudong Xie
- Department of Restorative Sciences, School of Dentistry, Texas A&M University, Dallas, TX, USA
| | - Jian Q Feng
- Department of Restorative Sciences, School of Dentistry, Texas A&M University, Dallas, TX, USA
| | - Qian Xu
- Department of Restorative Sciences, School of Dentistry, Texas A&M University, Dallas, TX, USA
| | - Yongbo Lu
- Department of Restorative Sciences, School of Dentistry, Texas A&M University, Dallas, TX, USA
| | - Robert E Hammer
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Carol A Wise
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, USA.,Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, USA.,McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX, USA.,Department of Orthopaedic Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Bruce Beutler
- Center for Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
19
|
Franco R, Rivas-Santisteban R, Navarro G, Pinna A, Reyes-Resina I. Genes Implicated in Familial Parkinson's Disease Provide a Dual Picture of Nigral Dopaminergic Neurodegeneration with Mitochondria Taking Center Stage. Int J Mol Sci 2021; 22:4643. [PMID: 33924963 PMCID: PMC8124903 DOI: 10.3390/ijms22094643] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/29/2021] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 12/13/2022] Open
Abstract
The mechanism of nigral dopaminergic neuronal degeneration in Parkinson's disease (PD) is unknown. One of the pathological characteristics of the disease is the deposition of α-synuclein (α-syn) that occurs in the brain from both familial and sporadic PD patients. This paper constitutes a narrative review that takes advantage of information related to genes (SNCA, LRRK2, GBA, UCHL1, VPS35, PRKN, PINK1, ATP13A2, PLA2G6, DNAJC6, SYNJ1, DJ-1/PARK7 and FBXO7) involved in familial cases of Parkinson's disease (PD) to explore their usefulness in deciphering the origin of dopaminergic denervation in many types of PD. Direct or functional interactions between genes or gene products are evaluated using the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) database. The rationale is to propose a map of the interactions between SNCA, the gene encoding for α-syn that aggregates in PD, and other genes, the mutations of which lead to early-onset PD. The map contrasts with the findings obtained using animal models that are the knockout of one of those genes or that express the mutated human gene. From combining in silico data from STRING-based assays with in vitro and in vivo data in transgenic animals, two likely mechanisms appeared: (i) the processing of native α-syn is altered due to the mutation of genes involved in vesicular trafficking and protein processing, or (ii) α-syn mutants alter the mechanisms necessary for the correct vesicular trafficking and protein processing. Mitochondria are a common denominator since both mechanisms require extra energy production, and the energy for the survival of neurons is obtained mainly from the complete oxidation of glucose. Dopamine itself can result in an additional burden to the mitochondria of dopaminergic neurons because its handling produces free radicals. Drugs acting on G protein-coupled receptors (GPCRs) in the mitochondria of neurons may hopefully end up targeting those receptors to reduce oxidative burden and increase mitochondrial performance. In summary, the analysis of the data of genes related to familial PD provides relevant information on the etiology of sporadic cases and might suggest new therapeutic approaches.
Collapse
Affiliation(s)
- Rafael Franco
- Department Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain; (R.F.); (R.R.-S.); (I.R.-R.)
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, 28031 Madrid, Spain;
| | - Rafael Rivas-Santisteban
- Department Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain; (R.F.); (R.R.-S.); (I.R.-R.)
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, 28031 Madrid, Spain;
| | - Gemma Navarro
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, 28031 Madrid, Spain;
- Department Biochemistry and Physiology, School of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain
| | - Annalisa Pinna
- National Research Council of Italy (CNR), Neuroscience Institute–Cagliari, Cittadella Universitaria, Blocco A, SP 8, Km 0.700, 09042 Monserrato (CA), Italy
| | - Irene Reyes-Resina
- Department Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain; (R.F.); (R.R.-S.); (I.R.-R.)
| |
Collapse
|
20
|
Brommage R, Jeter-Jones S, Xiong W, Liu J. MicroCT analyses of mouse femoral neck architecture. Bone 2021; 145:115040. [PMID: 31437568 DOI: 10.1016/j.bone.2019.115040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 12/07/2018] [Revised: 07/24/2019] [Accepted: 08/17/2019] [Indexed: 10/26/2022]
Abstract
Hip fractures at the femoral neck are a major cause of morbidity and mortality, but aside from biomechanical strength testing, little is known about femoral neck architecture in mice. Procedures were optimized to analyze high-resolution (6 μm voxel size) microCT scans of the mouse femoral neck to provide bone mass and architectural information. Similar to histomorphometric observations in rats, the boundary between cortical and trabecular bone is difficult to identify in the mouse femoral mid-neck and these compartments were not analyzed separately. Analyses included total area, mineralized bone area, and bone volume fraction (BV/TV). Femoral neck architecture varies in C57BL/6J, 129/SvEv and BALB/c mouse strains. Bone cross sectional area and BV/TV were low in Lrp5 but elevated in Sost gene knockout mice. Sfrp4 gene knockout resulted in high total area, normal bone area, low BV/TV and, as indicated by BS/BV values, greater trabecularization. Femoral neck BV/TV declined with age and ovariectomy, but increased with teriparatide treatment. These findings demonstrate that the architecture of the mouse femoral neck mimics phenotypes and treatment effects observed at other skeletal sites and is a relevant bone site for translational studies examining osteoporosis therapies.
Collapse
Affiliation(s)
- Robert Brommage
- Metabolism Research, Lexicon Pharmaceuticals, The Woodlands, TX, USA.
| | | | - Wendy Xiong
- Metabolism Research, Lexicon Pharmaceuticals, The Woodlands, TX, USA
| | - Jeff Liu
- Metabolism Research, Lexicon Pharmaceuticals, The Woodlands, TX, USA
| |
Collapse
|
21
|
Malik AU, Karapetsas A, Nirujogi RS, Mathea S, Chatterjee D, Pal P, Lis P, Taylor M, Purlyte E, Gourlay R, Dorward M, Weidlich S, Toth R, Polinski NK, Knapp S, Tonelli F, Alessi DR. Deciphering the LRRK code: LRRK1 and LRRK2 phosphorylate distinct Rab proteins and are regulated by diverse mechanisms. Biochem J 2021; 478:553-578. [PMID: 33459343 PMCID: PMC7886321 DOI: 10.1042/bcj20200937] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/25/2020] [Revised: 01/08/2021] [Accepted: 01/18/2021] [Indexed: 01/05/2023]
Abstract
Autosomal dominant mutations in LRRK2 that enhance kinase activity cause Parkinson's disease. LRRK2 phosphorylates a subset of Rab GTPases including Rab8A and Rab10 within its effector binding motif. Here, we explore whether LRRK1, a less studied homolog of LRRK2 that regulates growth factor receptor trafficking and osteoclast biology might also phosphorylate Rab proteins. Using mass spectrometry, we found that in LRRK1 knock-out cells, phosphorylation of Rab7A at Ser72 was most impacted. This residue lies at the equivalent site targeted by LRRK2 on Rab8A and Rab10. Accordingly, recombinant LRRK1 efficiently phosphorylated Rab7A at Ser72, but not Rab8A or Rab10. Employing a novel phospho-specific antibody, we found that phorbol ester stimulation of mouse embryonic fibroblasts markedly enhanced phosphorylation of Rab7A at Ser72 via LRRK1. We identify two LRRK1 mutations (K746G and I1412T), equivalent to the LRRK2 R1441G and I2020T Parkinson's mutations, that enhance LRRK1 mediated phosphorylation of Rab7A. We demonstrate that two regulators of LRRK2 namely Rab29 and VPS35[D620N], do not influence LRRK1. Widely used LRRK2 inhibitors do not inhibit LRRK1, but we identify a promiscuous inhibitor termed GZD-824 that inhibits both LRRK1 and LRRK2. The PPM1H Rab phosphatase when overexpressed dephosphorylates Rab7A. Finally, the interaction of Rab7A with its effector RILP is not affected by LRRK1 phosphorylation and we observe that maximal stimulation of the TBK1 or PINK1 pathway does not elevate Rab7A phosphorylation. Altogether, these findings reinforce the idea that the LRRK enzymes have evolved as major regulators of Rab biology with distinct substrate specificity.
Collapse
Affiliation(s)
- Asad U. Malik
- Medical Research Council (MRC) Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, U.K
| | - Athanasios Karapetsas
- Medical Research Council (MRC) Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, U.K
| | - Raja S. Nirujogi
- Medical Research Council (MRC) Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, U.K
| | - Sebastian Mathea
- Structural Genomics Consortium, Institute for Pharmaceutical Chemistry and Buchmann Institute for Molecular Life Sciences, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 9, D-60438 Frankfurt am Main, Germany
| | - Deep Chatterjee
- Structural Genomics Consortium, Institute for Pharmaceutical Chemistry and Buchmann Institute for Molecular Life Sciences, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 9, D-60438 Frankfurt am Main, Germany
| | - Prosenjit Pal
- Medical Research Council (MRC) Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, U.K
| | - Pawel Lis
- Medical Research Council (MRC) Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, U.K
| | - Matthew Taylor
- Medical Research Council (MRC) Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, U.K
| | - Elena Purlyte
- Medical Research Council (MRC) Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, U.K
| | - Robert Gourlay
- Medical Research Council (MRC) Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, U.K
| | - Mark Dorward
- Medical Research Council (MRC) Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, U.K
| | - Simone Weidlich
- Medical Research Council (MRC) Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, U.K
| | - Rachel Toth
- Medical Research Council (MRC) Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, U.K
| | - Nicole K. Polinski
- Michael J Fox Foundation for Parkinson's Research, Grand Central Station, PO Box 4777, New York, NY 10163, U.S.A
| | - Stefan Knapp
- Structural Genomics Consortium, Institute for Pharmaceutical Chemistry and Buchmann Institute for Molecular Life Sciences, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 9, D-60438 Frankfurt am Main, Germany
| | - Francesca Tonelli
- Medical Research Council (MRC) Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, U.K
| | - Dario R. Alessi
- Medical Research Council (MRC) Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, U.K
| |
Collapse
|
22
|
Chen Z, Xing W, Fan L. Chemical IN04 Inhibits the Kinase Domain not the ROC Domain of LRRK1: Results from Homology Modeling and Molecular Docking. Med Chem 2021; 17:1140-1150. [PMID: 32972350 PMCID: PMC7987913 DOI: 10.2174/1573406416666200924125620] [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] [Academic Contribution Register] [Received: 06/05/2020] [Revised: 08/17/2020] [Accepted: 08/17/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Bone loss is the most common reason for broken bones among the elderly. An ideal agent for the treatment of bone loss should have both osteoclast inhibitory and osteoblast stimulatory functions. Leucine-rich repeat kinase 1 (LRRK1) is a novel target for alternative antiresorptive drugs to treat osteoporosis and osteoporotic fractures. Recently a chemical IN04, Methyl 3-[({([5-(3,5-dimethoxyphenyl)-1,3,4-oxadiazol-2-yl]-thio}-acetyl)-amino]-benzoate, has been identified as a potential LRRK1 inhibitor. OBJECTIVE The aim of this work is to investigate how the chemical IN04 interacts with LRRK1 and inhibits its activity. METHODS A structural model of the LRRK1 kinase domain was constructed with SWISS-MODEL. The human protein kinase ROCO4 (PDB ID: 4YZN) was chosen as the template based on sequence homology, structural and phylogenetic analysis. In addition, a homology model of the LRRK1 ROC domain was also prepared based on the LRRK2 ROC domain structure (PDB ID: 2ZEJ). The interactions of IN04 with the active sites in the LRRK1 kinase domain and ROC domain were investigated by SwissDock. RESULTS IN04 was docked into the active site of the LRRK1 kinase domain with similar interactions as ATP comparable to the ligand bound to homologous kinases. Many rational binding modes of IN04 to LRRK1 kinase domain were investigated and the most likely binding pose containing multiple hydrogen bonds and a salt bridge was discovered. However, IN04 cannot fit into the GDP-binding site of the ROC domain. CONCLUSION Chemical IN04 inhibits LRRK1 by binding to the active site of the kinase domain but not the ROC domain.
Collapse
Affiliation(s)
- Zhenhang Chen
- Department of Biochemistry, University of California Riverside, Riverside, CA, USA
| | - Weirong Xing
- Department of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Li Fan
- Department of Biochemistry, University of California Riverside, Riverside, CA, USA
| |
Collapse
|
23
|
Abstract
The phenotypic trait of high bone mass (HBM) is an excellent example of the nexus between common and rare disease genetics. HBM may arise from carriage of many 'high bone mineral density [BMD]'-associated alleles, and certainly the genetic architecture of individuals with HBM is enriched with high BMD variants identified through genome-wide association studies of BMD. HBM may also arise as a monogenic skeletal disorder, due to abnormalities in bone formation, bone resorption, and/or bone turnover. Individuals with monogenic disorders of HBM usually, though not invariably, have other skeletal abnormalities (such as mandible enlargement) and thus are best regarded as having a skeletal dysplasia rather than just isolated high BMD. A binary etiological division of HBM into polygenic vs. monogenic, however, would be excessively simplistic: the phenotype of individuals carrying rare variants of large effect can still be modified by their common variant polygenic background, and by the environment. HBM disorders-whether predominantly polygenic or monogenic in origin-are not only interesting clinically and genetically: they provide insights into bone processes that can be exploited therapeutically, with benefits both for individuals with these rare bone disorders and importantly for the many people affected by the commonest bone disease worldwide-i.e., osteoporosis. In this review we detail the genetic architecture of HBM; we provide a conceptual framework for considering HBM in the clinical context; and we discuss monogenic and polygenic causes of HBM with particular emphasis on anabolic causes of HBM.
Collapse
Affiliation(s)
- Celia L. Gregson
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Emma L. Duncan
- Department of Twin Research & Genetic Epidemiology, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| |
Collapse
|
24
|
Blangy A, Bompard G, Guerit D, Marie P, Maurin J, Morel A, Vives V. The osteoclast cytoskeleton - current understanding and therapeutic perspectives for osteoporosis. J Cell Sci 2020; 133:133/13/jcs244798. [PMID: 32611680 DOI: 10.1242/jcs.244798] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/30/2022] Open
Abstract
Osteoclasts are giant multinucleated myeloid cells specialized for bone resorption, which is essential for the preservation of bone health throughout life. The activity of osteoclasts relies on the typical organization of osteoclast cytoskeleton components into a highly complex structure comprising actin, microtubules and other cytoskeletal proteins that constitutes the backbone of the bone resorption apparatus. The development of methods to differentiate osteoclasts in culture and manipulate them genetically, as well as improvements in cell imaging technologies, has shed light onto the molecular mechanisms that control the structure and dynamics of the osteoclast cytoskeleton, and thus the mechanism of bone resorption. Although essential for normal bone physiology, abnormal osteoclast activity can cause bone defects, in particular their hyper-activation is commonly associated with many pathologies, hormonal imbalance and medical treatments. Increased bone degradation by osteoclasts provokes progressive bone loss, leading to osteoporosis, with the resulting bone frailty leading to fractures, loss of autonomy and premature death. In this context, the osteoclast cytoskeleton has recently proven to be a relevant therapeutic target for controlling pathological bone resorption levels. Here, we review the present knowledge on the regulatory mechanisms of the osteoclast cytoskeleton that control their bone resorption activity in normal and pathological conditions.
Collapse
Affiliation(s)
- Anne Blangy
- Centre de Recherche de Biologie Cellulaire de Montpellier (CRBM), Montpellier Univ., CNRS, 34000 Montpellier, France
| | - Guillaume Bompard
- Centre de Recherche de Biologie Cellulaire de Montpellier (CRBM), Montpellier Univ., CNRS, 34000 Montpellier, France
| | - David Guerit
- Centre de Recherche de Biologie Cellulaire de Montpellier (CRBM), Montpellier Univ., CNRS, 34000 Montpellier, France
| | - Pauline Marie
- Centre de Recherche de Biologie Cellulaire de Montpellier (CRBM), Montpellier Univ., CNRS, 34000 Montpellier, France
| | - Justine Maurin
- Centre de Recherche de Biologie Cellulaire de Montpellier (CRBM), Montpellier Univ., CNRS, 34000 Montpellier, France
| | - Anne Morel
- Centre de Recherche de Biologie Cellulaire de Montpellier (CRBM), Montpellier Univ., CNRS, 34000 Montpellier, France
| | - Virginie Vives
- Centre de Recherche de Biologie Cellulaire de Montpellier (CRBM), Montpellier Univ., CNRS, 34000 Montpellier, France
| |
Collapse
|
25
|
Howaldt A, Hennig AF, Rolvien T, Rössler U, Stelzer N, Knaus A, Böttger S, Zustin J, Geißler S, Oheim R, Amling M, Howaldt HP, Kornak U. Adult Osteosclerotic Metaphyseal Dysplasia With Progressive Osteonecrosis of the Jaws and Abnormal Bone Resorption Pattern Due to a LRRK1 Splice Site Mutation. J Bone Miner Res 2020; 35:1322-1332. [PMID: 32119750 DOI: 10.1002/jbmr.3995] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 10/16/2019] [Revised: 02/14/2020] [Accepted: 02/27/2020] [Indexed: 12/15/2022]
Abstract
Osteosclerotic metaphyseal dysplasia (OSMD) is a rare autosomal recessive sclerosing skeletal dysplasia. We report on a 34-year-old patient with sandwich vertebrae, platyspondyly, osteosclerosis of the tubular bones, pathologic fractures, and anemia. In the third decade, he developed osteonecrosis of the jaws, which was progressive in spite of repeated surgical treatment over a period of 11 years. An iliac crest bone biopsy revealed the presence of hypermineralized cartilage remnants, large multinucleated osteoclasts with abnormal morphology, and inadequate bone resorption typical for osteoclast-rich osteopetrosis. After exclusion of mutations in TCIRG1 and CLCN7 we performed trio-based exome sequencing. The novel homozygous splice-site mutation c.261G>A in the gene LRRK1 was found and co-segregated with the phenotype in the family. cDNA sequencing showed nearly complete skipping of exon 3 leading to a frameshift (p.Ala34Profs*33). Osteoclasts differentiated from the patient's peripheral blood monocytes were extremely large. Instead of resorption pits these cells were only capable of superficial erosion. Phosphorylation of L-plastin at position Ser5 was strongly reduced in patient-derived osteoclasts showing a loss of function of the mutated LRRK1 kinase protein. Our analysis indicates a strong overlap of LRRK1-related OSMD with other forms of intermediate osteopetrosis, but an exceptional abnormality of osteoclast resorption. Like in other osteoclast pathologies an increased risk for progressive osteonecrosis of the jaws should be considered in OSMD, an intermediate form of osteopetrosis. © 2020 The Authors. Journal of Bone and Mineral Research published by American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Antonia Howaldt
- Institut für Medizinische Genetik und Humangenetik, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,BIH Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Anna Floriane Hennig
- Institut für Medizinische Genetik und Humangenetik, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,BIH Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Freie Universität Berlin, Berlin, Germany
| | - Tim Rolvien
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Orthopedics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Uta Rössler
- Institut für Medizinische Genetik und Humangenetik, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,BIH Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Nina Stelzer
- Institut für Medizinische Genetik und Humangenetik, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,BIH Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Alexej Knaus
- Institute for Genomic Statistics and Bioinformatics, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Sebastian Böttger
- Department for Maxillo Facial Surgery, Justus Liebig University Gießen, Gießen, Germany
| | - Jozef Zustin
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sven Geißler
- BIH Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ralf Oheim
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hans-Peter Howaldt
- Department for Maxillo Facial Surgery, Justus Liebig University Gießen, Gießen, Germany
| | - Uwe Kornak
- Institut für Medizinische Genetik und Humangenetik, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,BIH Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Max Planck Institute for Molecular Genetics, Berlin, Germany.,Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
26
|
Zhang Y, Cui Y, Wang L, Han J. Autophagy promotes osteoclast podosome disassembly and cell motility athrough the interaction of kindlin3 with LC3. Cell Signal 2020; 67:109505. [DOI: 10.1016/j.cellsig.2019.109505] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/22/2019] [Revised: 12/16/2019] [Accepted: 12/16/2019] [Indexed: 12/21/2022]
|
27
|
Miryounesi M, Nikfar A, Changi‐Ashtiani M, Shahrooei M, Dinmohammadi H, Shahani T, Zarvandi S, Bahrami T, Momenilandi M, Rokni‐Zadeh H. A novel homozygous
LRRK1
stop gain mutation in a patient suspected with osteosclerotic metaphyseal dysplasia. Ann Hum Genet 2019; 84:102-106. [DOI: 10.1111/ahg.12352] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/17/2018] [Revised: 07/25/2019] [Accepted: 08/27/2019] [Indexed: 01/06/2023]
Affiliation(s)
- Mohammad Miryounesi
- Genomic Research Center Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Ali Nikfar
- Department of Genetics and Molecular Medicine, School of Medicine Zanjan University of Medical Sciences (ZUMS) Zanjan Iran
| | - Majid Changi‐Ashtiani
- School of Mathematics Institute for Research in Fundamental Sciences (IPM) Tehran Iran
| | - Mohammad Shahrooei
- Department of Microbiology and Immunology, Laboratory of Clinical Bacteriology and Mycology KU Leuven Leuven Belgium
- Specialized Immunology Laboratory of Dr. Shahrooei Ahvaz Iran
| | - Hossein Dinmohammadi
- Department of Genetics and Molecular Medicine, School of Medicine Zanjan University of Medical Sciences (ZUMS) Zanjan Iran
| | - Tina Shahani
- Department of Genetics and Molecular Medicine, School of Medicine Zanjan University of Medical Sciences (ZUMS) Zanjan Iran
| | - Samira Zarvandi
- Department of Medical Biotechnology, School of Medicine Zanjan University of Medical Sciences (ZUMS) Zanjan Iran
| | - Tahereh Bahrami
- Department of Medical Biotechnology, School of Medicine Zanjan University of Medical Sciences (ZUMS) Zanjan Iran
| | | | - Hassan Rokni‐Zadeh
- Department of Medical Biotechnology, School of Medicine Zanjan University of Medical Sciences (ZUMS) Zanjan Iran
| |
Collapse
|
28
|
Xing W, Godwin C, Pourteymoor S, Mohan S. Conditional disruption of the osterix gene in chondrocytes during early postnatal growth impairs secondary ossification in the mouse tibial epiphysis. Bone Res 2019; 7:24. [PMID: 31646014 PMCID: PMC6804621 DOI: 10.1038/s41413-019-0064-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/05/2018] [Revised: 06/04/2019] [Accepted: 06/19/2019] [Indexed: 12/12/2022] Open
Abstract
In our previous studies, we have found that the prepubertal increase in thyroid hormone levels induces osterix (Osx) signaling in hypertrophic chondrocytes to transdifferentiate them into osteoblasts. To test if Osx expressed in chondrocytes directly contributes to transdifferentiation and secondary ossification, we generated Osx flox/flox ; Col2-Cre-ERT2 mice and knocked out Osx with a single injection of tamoxifen at postnatal day (P) 3 prior to evaluation of the epiphyseal bone phenotype by µCT, histology, and immunohistochemistry (IHC) at P21. Vehicle (oil)-treated Osx flox/flox ; Col2-Cre-ERT2 and tamoxifen-treated, Cre-negative Osx flox/flox mice were used as controls. µCT analysis of tibial epiphyses revealed that trabecular bone mass was reduced by 23% in the Osx conditional knockout (cKO) compared with control mice. Trabecular number and thickness were reduced by 28% and 8%, respectively, while trabecular separation was increased by 24% in the cKO mice. Trichrome staining of longitudinal sections of tibial epiphyses showed that bone area and bone area adjusted for total area were decreased by 22% and 18%, respectively. IHC studies revealed the presence of abundant Osx-expressing prehypertrophic chondrocytes in the epiphyses of control mice at P10, but not in the cKO mice. Furthermore, expression levels of MMP13, COL10, ALP, and BSP were considerably reduced in the epiphyses of cKO mice. We also found that Osx overexpression in ATDC5 chondrocytes increased expression of Col10, Mmp13, Alp, and Bsp. Our data indicate that Osx expressed in chondrocytes plays a significant role in secondary ossification by regulating expression of genes involved in chondrocyte hypertrophy and osteoblast transdifferentiation.
Collapse
Affiliation(s)
- Weirong Xing
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, CA 92357 USA
- Department of Medicine, Loma Linda University, Loma Linda, CA 92357 USA
| | - Catrina Godwin
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, CA 92357 USA
| | - Sheila Pourteymoor
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, CA 92357 USA
| | - Subburaman Mohan
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, CA 92357 USA
- Department of Medicine, Loma Linda University, Loma Linda, CA 92357 USA
- Department of Orthopedics, Loma Linda University, Loma Linda, CA 92357 USA
- Department of Biochemistry, Loma Linda University, Loma Linda, CA 92357 USA
| |
Collapse
|
29
|
Si M, Zeng C, Goodluck H, Shen S, Mohan S, Xing W. A small molecular inhibitor of LRRK1 identified by homology modeling and virtual screening suppresses osteoclast function, but not osteoclast differentiation, in vitro. Aging (Albany NY) 2019; 11:3250-3261. [PMID: 31113907 PMCID: PMC6555463 DOI: 10.18632/aging.101977] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/28/2019] [Accepted: 05/12/2019] [Indexed: 01/31/2023]
Abstract
We used TGFβ activation kinase 1 as a template to build a 3D structure of the human LRRK1 kinase domain (hLRRK1 KD) and performed small molecule docking. One of the chemicals (IN04) that docked into the pocket was chosen for evaluation of biological effects on osteoclasts (OCs) in vitro. INO4 at 16 nM completely blocked ATP binding to hLRRK1 KD in an in vitro pulldown assay. In differentiation and pit assays, while the number of OCs on bone slices were comparable for OCs treated with IN04 and DMSO, IN04 treatment of OCs significantly impaired their ability to resorb bone. The area of pits on bone slices was reduced by 43% at 5 μM and 83% at 10 μM as compared to DMSO. Individual pits appeared smaller and shallower. F-actin staining revealed that DMSO-treated OCs displayed clear actin rings, and F-actin forms a peripheral sealing zone. By contrast, IN04-treated OCs showed disarranged F-actin in the cytoplasm, and F-actin failed to form a sealing zone on bone slices. IN04 treatment had no effects on OC-derived coupling factor production nor on osteoblast nodule formation. Our data indicate IN04 is a potent inhibitor of LRRK1, suppressing OC function with no effect on OC formation.
Collapse
Affiliation(s)
- Mingjue Si
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, Loma Linda 92357, CA, USA
- Department of Medicine, Loma Linda University, Loma Linda 92350, CA, USA
- Equal contribution
| | - Canjun Zeng
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, Loma Linda 92357, CA, USA
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Equal contribution
| | - Helen Goodluck
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, Loma Linda 92357, CA, USA
- Equal contribution
| | - Sandi Shen
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, Loma Linda 92357, CA, USA
- Department of Medicine, Loma Linda University, Loma Linda 92350, CA, USA
| | - Subburaman Mohan
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, Loma Linda 92357, CA, USA
- Department of Medicine, Loma Linda University, Loma Linda 92350, CA, USA
| | - Weirong Xing
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, Loma Linda 92357, CA, USA
- Department of Medicine, Loma Linda University, Loma Linda 92350, CA, USA
| |
Collapse
|
30
|
Brommage R, Powell DR, Vogel P. Predicting human disease mutations and identifying drug targets from mouse gene knockout phenotyping campaigns. Dis Model Mech 2019; 12:dmm038224. [PMID: 31064765 PMCID: PMC6550044 DOI: 10.1242/dmm.038224] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/24/2022] Open
Abstract
Two large-scale mouse gene knockout phenotyping campaigns have provided extensive data on the functions of thousands of mammalian genes. The ongoing International Mouse Phenotyping Consortium (IMPC), with the goal of examining all ∼20,000 mouse genes, has examined 5115 genes since 2011, and phenotypic data from several analyses are available on the IMPC website (www.mousephenotype.org). Mutant mice having at least one human genetic disease-associated phenotype are available for 185 IMPC genes. Lexicon Pharmaceuticals' Genome5000™ campaign performed similar analyses between 2000 and the end of 2008 focusing on the druggable genome, including enzymes, receptors, transporters, channels and secreted proteins. Mutants (4654 genes, with 3762 viable adult homozygous lines) with therapeutically interesting phenotypes were studied extensively. Importantly, phenotypes for 29 Lexicon mouse gene knockouts were published prior to observations of similar phenotypes resulting from homologous mutations in human genetic disorders. Knockout mouse phenotypes for an additional 30 genes mimicked previously published human genetic disorders. Several of these models have helped develop effective treatments for human diseases. For example, studying Tph1 knockout mice (lacking peripheral serotonin) aided the development of telotristat ethyl, an approved treatment for carcinoid syndrome. Sglt1 (also known as Slc5a1) and Sglt2 (also known as Slc5a2) knockout mice were employed to develop sotagliflozin, a dual SGLT1/SGLT2 inhibitor having success in clinical trials for diabetes. Clinical trials evaluating inhibitors of AAK1 (neuropathic pain) and SGLT1 (diabetes) are underway. The research community can take advantage of these unbiased analyses of gene function in mice, including the minimally studied 'ignorome' genes.
Collapse
Affiliation(s)
- Robert Brommage
- Department of Metabolism Research, Lexicon Pharmaceuticals, 8800 Technology Forest Place, The Woodlands, TX 77381, USA
| | - David R Powell
- Department of Metabolism Research, Lexicon Pharmaceuticals, 8800 Technology Forest Place, The Woodlands, TX 77381, USA
| | - Peter Vogel
- St. Jude Children's Research Hospital, Pathology, MS 250, Room C5036A, 262 Danny Thomas Place, Memphis, TN 38105, USA
| |
Collapse
|
31
|
Brommage R, Liu J, Vogel P, Mseeh F, Thompson AY, Potter DG, Shadoan MK, Hansen GM, Jeter-Jones S, Cui J, Bright D, Bardenhagen JP, Doree DD, Movérare-Skrtic S, Nilsson KH, Henning P, Lerner UH, Ohlsson C, Sands AT, Tarver JE, Powell DR, Zambrowicz B, Liu Q. NOTUM inhibition increases endocortical bone formation and bone strength. Bone Res 2019; 7:2. [PMID: 30622831 PMCID: PMC6323125 DOI: 10.1038/s41413-018-0038-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/13/2018] [Revised: 09/21/2018] [Accepted: 10/15/2018] [Indexed: 12/16/2022] Open
Abstract
The disability, mortality and costs caused by non-vertebral osteoporotic fractures are enormous. Existing osteoporosis therapies are highly effective at reducing vertebral but not non-vertebral fractures. Cortical bone is a major determinant of non-vertebral bone strength. To identify novel osteoporosis drug targets, we phenotyped cortical bone of 3 366 viable mouse strains with global knockouts of druggable genes. Cortical bone thickness was substantially elevated in Notum−/− mice. NOTUM is a secreted WNT lipase and we observed high NOTUM expression in cortical bone and osteoblasts but not osteoclasts. Three orally active small molecules and a neutralizing antibody inhibiting NOTUM lipase activity were developed. They increased cortical bone thickness and strength at multiple skeletal sites in both gonadal intact and ovariectomized rodents by stimulating endocortical bone formation. Thus, inhibition of NOTUM activity is a potential novel anabolic therapy for strengthening cortical bone and preventing non-vertebral fractures. NOTUM is an enzyme that inactivates WNT proteins (which play a key role in early tissue development), and inhibiting NOTUM has been found to increase the formation of endocortical bone (within the cortex, the hard exterior of bone) and enhance bone strength. Existing therapies for osteoporosis (condition causing bone to become weak and brittle) are effective in reducing vertebral, but not non-vertebral, fractures. A team headed by Robert Brommage at Lexicon Pharmaceuticals, Texas aimed to identify novel osteoporosis drug targets in mice. Following inhibition of NOTUM activity, the authors observed increased cortical bone thickness and strength at multiple skeletal sites through stimulation of endocortical bone formation. The team concluded that inhibiting NOTUM activity has good potential as a new therapeutic strategy and could be beneficial in preventing non-vertebral osteoporotic fractures.
Collapse
Affiliation(s)
- Robert Brommage
- 1Lexicon Pharmaceuticals, The Woodlands, TX USA.,3Present Address: Centre for Bone and Arthritis Research, University of Gothenburg, Gothenburg, Sweden
| | - Jeff Liu
- 1Lexicon Pharmaceuticals, The Woodlands, TX USA.,4Present Address: Biogen, Cambridge, MA, USA
| | - Peter Vogel
- 1Lexicon Pharmaceuticals, The Woodlands, TX USA.,5Present Address: St. Jude Children's Research Hospital, Memphis, TN USA
| | - Faika Mseeh
- 1Lexicon Pharmaceuticals, The Woodlands, TX USA.,6Present Address: MD Anderson Cancer Center, Houston, TX USA
| | | | - David G Potter
- 1Lexicon Pharmaceuticals, The Woodlands, TX USA.,Present Address: Bethyl Laboratories, Montgomery, TX USA
| | - Melanie K Shadoan
- 1Lexicon Pharmaceuticals, The Woodlands, TX USA.,8Present Address: Merck, Rahway, NJ USA
| | - Gwenn M Hansen
- 1Lexicon Pharmaceuticals, The Woodlands, TX USA.,Present Address: Nurix, San Francisco, CA USA
| | - Sabrina Jeter-Jones
- 1Lexicon Pharmaceuticals, The Woodlands, TX USA.,6Present Address: MD Anderson Cancer Center, Houston, TX USA
| | - Jie Cui
- 1Lexicon Pharmaceuticals, The Woodlands, TX USA.,Present Address: Wntrix, Houston, TX USA
| | - Dawn Bright
- 1Lexicon Pharmaceuticals, The Woodlands, TX USA
| | - Jennifer P Bardenhagen
- 1Lexicon Pharmaceuticals, The Woodlands, TX USA.,6Present Address: MD Anderson Cancer Center, Houston, TX USA
| | - Deon D Doree
- 1Lexicon Pharmaceuticals, The Woodlands, TX USA.,11Present Address: PRA Health Sciences, Raleigh, NC USA
| | - Sofia Movérare-Skrtic
- 2Centre for Bone and Arthritis Research, Institute of Medicine, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Karin H Nilsson
- 2Centre for Bone and Arthritis Research, Institute of Medicine, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Petra Henning
- 2Centre for Bone and Arthritis Research, Institute of Medicine, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Ulf H Lerner
- 2Centre for Bone and Arthritis Research, Institute of Medicine, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Claes Ohlsson
- 2Centre for Bone and Arthritis Research, Institute of Medicine, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Arthur T Sands
- 1Lexicon Pharmaceuticals, The Woodlands, TX USA.,Present Address: Nurix, San Francisco, CA USA
| | - James E Tarver
- 1Lexicon Pharmaceuticals, The Woodlands, TX USA.,12Present Address: University of Pennsylvania, Philadelphia, PA USA
| | | | - Brian Zambrowicz
- 1Lexicon Pharmaceuticals, The Woodlands, TX USA.,13Present Address: Regeneron Pharmaceuticals, Tarrytown, NY USA
| | - Qingyun Liu
- 1Lexicon Pharmaceuticals, The Woodlands, TX USA.,14Present Address: University of Texas, Houston, TX USA
| |
Collapse
|
32
|
Si M, Goodluck H, Zeng C, Pan S, Todd EM, Morley SC, Qin X, Mohan S, Xing W. LRRK1 regulation of actin assembly in osteoclasts involves serine 5 phosphorylation of L-plastin. J Cell Biochem 2018; 119:10351-10357. [PMID: 30136304 PMCID: PMC6218268 DOI: 10.1002/jcb.27377] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/28/2018] [Accepted: 07/02/2018] [Indexed: 12/11/2022]
Abstract
Mice with disruption of Lrrk1 and patients with nonfunctional mutant Lrrk1 exhibit severe osteopetrosis phenotypes because of osteoclast cytoskeletal dysfunction. To understand how Lrrk1 regulates osteoclast function by modulating cytoskeleton rearrangement, we examined the proteins that are differentially phosphorylated in wild‐type mice and Lrrk1‐deficient osteoclasts by metal affinity purification coupled liquid chromatography/mass spectrometry (LC/MS) analyses. One of the candidates that we identified by LC/MS is L‐plastin, an actin bundling protein. We found that phosphorylation of L‐plastin at serine (Ser) residues 5 was present in wild‐type osteoclasts but not in Lrrk1‐deficient cells. Western blot analyses with antibodies specific for Ser5 phosphorylated L‐plastin confirmed the reduced L‐plastin Ser5 phosphorylation in Lrrk1 knockout (KO) osteoclasts. micro computed tomography (Micro‐CT) analyses revealed that the trabecular bone volume of the distal femur was increased by 27% in the 16 to 21‐week‐old L‐plastin KO females as compared with the wild‐type control mice. The ratio of bone volume to tissue volume and connectivity density were increased by 44% and 47% (both P < 0.05), respectively, in L‐plastin KO mice. Our data suggest that targeted disruption of L‐plastin increases trabecular bone volume, and phosphorylation of Ser5 in L‐plastin in the Lrrk1 signaling pathway may in part contribute to actin assembly in mature osteoclasts.
Collapse
Affiliation(s)
- Mingjue Si
- Department of Radiology, Shanghai Ninth People's Hospital, Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China.,Department of Medicine, Loma Linda University, Loma Linda, California
| | - Helen Goodluck
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, Loma Linda, California
| | - Canjun Zeng
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, Loma Linda, California.,Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Songqin Pan
- Proteomics Core Facility, University of California, Riverside, California
| | - Elizabeth M Todd
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - Sharon Celeste Morley
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Xuezhong Qin
- Department of Medicine, Loma Linda University, Loma Linda, California.,Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, Loma Linda, California
| | - Subburaman Mohan
- Department of Medicine, Loma Linda University, Loma Linda, California.,Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, Loma Linda, California
| | - Weirong Xing
- Department of Medicine, Loma Linda University, Loma Linda, California.,Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, Loma Linda, California.,Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
33
|
Qing R, Huang Z, Tang Y, Xiang Q, Yang F. Cordycepin negatively modulates lipopolysaccharide-induced cytokine production by up-regulation of heme oxygenase-1. Int Immunopharmacol 2017; 47:20-27. [PMID: 28351780 DOI: 10.1016/j.intimp.2017.03.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/07/2016] [Revised: 02/19/2017] [Accepted: 03/01/2017] [Indexed: 12/12/2022]
Abstract
AIMS The present study is to investigate the effect of cordycepin on the expression of heme oxygenase-1 (HO-1) in lipopolysaccharide (LPS)-activated microphages, as well as its mechanism of action. METHODS Mouse RAW264.7 cells were treated with different concentrations of cordycepin for 0-16h. Western blotting was used to determine the expression of HO-1 and the phosphorylation of c-Src and the p47phox subunit of NADPH oxidase. Intracellular reactive oxygen species (ROS) level was determined using H2DCFDA as fluorescent probe. Laser-scanning confocal microscopy was used to visualize the nuclear translocation of NF-E2-related factor 2 (Nrf2). Enzyme-linked immunosorbent assay was performed to measure the inhibitory effect of cordycepin on LPS-induced secretion of tumor necrosis factor-α and interleukin-6. RESULTS Cordycepin induced the phosphorylation of c-Src and p47phox subunit of NADPH oxidase in RAW264.7 cells. Cordycepin increased the secretion of ROS by activating NADPH oxidase. In addition, cordycepin enhanced the expression of HO-1 in RAW264.7 cells in both dose- and time-dependent manners. Of note, elevated HO-1 expression induced by cordycepin treatment was regulated by c-Src/NADPH oxidase/ROS pathway. HO-1 expression induced by cordycepin was dependent on the activation of Nrf2, which was regulated by c-Src/NADPH oxidase/ROS. Cordycepin reduced LPS-induced secretion of proinflammatory cytokines through up-regulation of HO-1. CONCLUSION The present study demonstrates that cordycepin induces the expression of HO-1 in RAW264.7 cells via c-Src/NADPH oxidase/ROS/Nrf2 pathway, and plays an anti-inflammatory role by inhibiting the secretion of cytokines from macrophages.
Collapse
Affiliation(s)
- Rui Qing
- Division of Pathogenic Biology, Department of Laboratory Medicine, Shaoyang University, Shaoyang, PR China
| | - Zezhi Huang
- Division of Pathogenic Biology, Department of Laboratory Medicine, Shaoyang University, Shaoyang, PR China
| | - Yufei Tang
- Division of Pathogenic Biology, Department of Laboratory Medicine, Shaoyang University, Shaoyang, PR China
| | - Qingke Xiang
- Division of Pathogenic Biology, Department of Laboratory Medicine, Shaoyang University, Shaoyang, PR China
| | - Fan Yang
- Department of Basic Medicine, Xiangnan University, Chenzhou, PR China.
| |
Collapse
|
34
|
Xing WR, Goodluck H, Zeng C, Mohan S. Role and mechanism of action of leucine-rich repeat kinase 1 in bone. Bone Res 2017; 5:17003. [PMID: 28326224 PMCID: PMC5348726 DOI: 10.1038/boneres.2017.3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/14/2016] [Revised: 12/06/2016] [Accepted: 12/13/2016] [Indexed: 12/13/2022] Open
Abstract
Leucine-rich repeat kinase 1 (LRRK1) plays a critical role in regulating cytoskeletal organization, osteoclast activity, and bone resorption with little effect on bone formation parameters. Deficiency of Lrrk1 in mice causes a severe osteopetrosis in the metaphysis of the long bones and vertebrae bones, which makes LRRK1 an attractive alternative drug target for the treatment of osteoporosis and other high-turnover bone diseases. This review summarizes recent advances on the functions of the Lrrk1-related family members, Lrrk1 deficiency-induced skeletal phenotypes, LRRK1 structure–function, potential biological substrates and interacting proteins, and the mechanisms of LRRK1 action in osteoclasts.
Collapse
Affiliation(s)
- Weirong R Xing
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, Loma Linda, CA, USA; Department of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Helen Goodluck
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center , Loma Linda, CA, USA
| | - Canjun Zeng
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, Loma Linda, CA, USA; Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Subburaman Mohan
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, Loma Linda, CA, USA; Department of Medicine, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
35
|
Pathogenic LRRK2 variants are gain-of-function mutations that enhance LRRK2-mediated repression of β-catenin signaling. Mol Neurodegener 2017; 12:9. [PMID: 28103901 PMCID: PMC5248453 DOI: 10.1186/s13024-017-0153-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/18/2016] [Accepted: 01/06/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND LRRK2 mutations and risk variants increase susceptibility to inherited and idiopathic Parkinson's disease, while recent studies have identified potential protective variants. This, and the fact that LRRK2 mutation carriers develop symptoms and brain pathology almost indistinguishable from idiopathic Parkinson's disease, has led to enormous interest in this protein. LRRK2 has been implicated in a range of cellular events, but key among them is canonical Wnt signalling, which results in increased levels of transcriptionally active β-catenin. This pathway is critical for the development and survival of the midbrain dopaminergic neurones typically lost in Parkinson's disease. METHODS Here we use Lrrk2 knockout mice and fibroblasts to investigate the effect of loss of Lrrk2 on canonical Wnt signalling in vitro and in vivo. Micro-computed tomography was used to study predicted tibial strength, while pulldown assays were employed to measure brain β-catenin levels. A combination of luciferase assays, immunofluorescence and co-immunoprecipitation were performed to measure canonical Wnt activity and investigate the relationship between LRRK2 and β-catenin. TOPflash assays are also used to study the effects of LRRK2 kinase inhibition and pathogenic and protective LRRK2 mutations on Wnt signalling. Data were tested by Analysis of Variance. RESULTS Loss of Lrrk2 causes a dose-dependent increase in the levels of transcriptionally active β-catenin in the brain, and alters tibial bone architecture, decreasing the predicted risk of fracture. Lrrk2 knockout cells display increased TOPflash and Axin2 promoter activities, both basally and following Wnt activation. Consistently, over-expressed LRRK2 was found to bind β-catenin and repress TOPflash activation. Some pathogenic LRRK2 mutations and risk variants further suppressed TOPflash, whereas the protective R1398H variant increased Wnt signalling activity. LRRK2 kinase inhibitors affected canonical Wnt signalling differently due to off-targeting; however, specific LRRK2 inhibition reduced canonical Wnt signalling similarly to pathogenic mutations. CONCLUSIONS Loss of LRRK2 causes increased canonical Wnt activity in vitro and in vivo. In agreement, over-expressed LRRK2 binds and represses β-catenin, suggesting LRRK2 may act as part of the β-catenin destruction complex. Since some pathogenic LRRK2 mutations enhance this effect while the protective R1398H variant relieves it, our data strengthen the notion that decreased canonical Wnt activity is central to Parkinson's disease pathogenesis.
Collapse
|
36
|
Identification of a novel LRRK1 mutation in a family with osteosclerotic metaphyseal dysplasia. J Hum Genet 2016; 62:437-441. [PMID: 27829680 DOI: 10.1038/jhg.2016.136] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/23/2016] [Revised: 10/06/2016] [Accepted: 10/07/2016] [Indexed: 12/22/2022]
Abstract
Osteosclerotic metaphyseal dysplasia (OSMD) is a rare skeletal dysplasia characterized by osteosclerotic metaphyses with osteopenic diaphyses of the long tubular bones. Our previous study identified a homozygous elongation mutation in leucine-rich repeat kinase 1 gene (LRRK1) in a patient with OSMD and showed that Lrrk1 knockout mice exhibited phenotypic similarity with OSMD. Here we report a second LRRK1 mutation in Indian sibs with OSMD. They had homozygous mutation (c.5971_5972insG) that produces an elongated mutant protein (p.A1991Gfs*31) similar to the first case. The sibs had normal stature, normal intelligence and recurrent fractures. The common radiographic feature was asymmetric and variable sclerosis of vertebral end plates, pelvic margin and metaphyses of tubular bones. One of the sibs had facial dysmorphisms, dentine abnormalities and acro-osteolysis. A comparison between the three OSMD cases with LRRK1 mutations with different ages suggested that the sclerotic lesions resolved with age. Our findings further support that LRRK1 would cause a subset of OSMD cases.
Collapse
|
37
|
Zeng C, Goodluck H, Qin X, Liu B, Mohan S, Xing W. Leucine-rich repeat kinase-1 regulates osteoclast function by modulating RAC1/Cdc42 Small GTPase phosphorylation and activation. Am J Physiol Endocrinol Metab 2016; 311:E772-E780. [PMID: 27600824 PMCID: PMC5241559 DOI: 10.1152/ajpendo.00189.2016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 05/10/2016] [Accepted: 08/26/2016] [Indexed: 11/24/2022]
Abstract
Leucine-rich repeat kinase-1 (Lrrk1) consists of ankyrin repeats (ANK), leucine-rich repeats (LRR), a GTPase-like domain of Roc (ROC), a COR domain, a serine/threonine kinase domain (KD), and WD40 repeats (WD40). Previous studies have revealed that knockout (KO) of Lrrk1 in mice causes severe osteopetrosis, and a human mutation of Lrrk1 leads to osteosclerotic metaphysial dysplasia. The molecular mechanism by which Lrrk1 regulates osteoclast function is unknown. In this study, we generated a series of Lrrk1 mutants and evaluated their ability to rescue defective bone resorption in Lrrk1-deficient osteoclasts by use of pit formation assays. Overexpression of Lrrk1 or LRR-truncated Lrrk1, but not ANK-truncated Lrrk1, WD40-truncated Lrrk1, Lrrk1-KD, or K651A mutant Lrrk1, rescued bone resorption function of Lrrk1 KO osteoclasts. We next examined whether RAC1/Cdc42 small GTPases are direct substrates of Lrrk1 in osteoclasts. Western blot and pull-down assays revealed that Lrrk1 deficiency in osteoclasts resulted in reduced phosphorylation and activation of RAC1/Cdc42. In vitro kinase assays confirmed that recombinant Lrrk1 phosphorylated RAC1-GST protein, and immunoprecipitation showed that the interaction of Lrrk1 with RAC1 occurred within 10 min after RANKL treatment. Overexpression of constitutively active Q61L RAC1 partially rescued the resorptive function of Lrrk1-deficient osteoclasts. Furthermore, lack of Lrrk1 in osteoclasts led to reduced autophosphorylation of p21 protein-activated kinase-1 at Ser144, catalyzed by RAC1/Cdc42 binding and activation. Our data indicate that Lrrk1 regulates osteoclast function by directly modulating phosphorylation and activation of small GTPase RAC1/Cdc42 and that its function depends on ANK, ROC, WD40, and kinase domains.
Collapse
Affiliation(s)
- Canjun Zeng
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial Veterans Affairs Medical Center, Loma Linda, California; Department of Orthopedics, The Third Affiliated Hospital Of Southern Medical University, Guangzhou, China
| | - Helen Goodluck
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial Veterans Affairs Medical Center, Loma Linda, California
| | - Xuezhong Qin
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial Veterans Affairs Medical Center, Loma Linda, California; Department of Medicine, Loma Linda University, Loma Linda, California
| | - Bo Liu
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial Veterans Affairs Medical Center, Loma Linda, California; Department of Orthopedics, The Third Xiangya Hosptial, Central South University, Changsha, Hunan, China
| | - Subburaman Mohan
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial Veterans Affairs Medical Center, Loma Linda, California; Department of Medicine, Loma Linda University, Loma Linda, California
| | - Weirong Xing
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial Veterans Affairs Medical Center, Loma Linda, California; Department of Medicine, Loma Linda University, Loma Linda, California;
| |
Collapse
|
38
|
Xing W, Aghajanian P, Goodluck H, Kesavan C, Cheng S, Pourteymoor S, Watt H, Alarcon C, Mohan S. Thyroid hormone receptor-β1 signaling is critically involved in regulating secondary ossification via promoting transcription of the Ihh gene in the epiphysis. Am J Physiol Endocrinol Metab 2016; 310:E846-54. [PMID: 27026086 PMCID: PMC4895449 DOI: 10.1152/ajpendo.00541.2015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 12/31/2015] [Accepted: 03/17/2016] [Indexed: 12/18/2022]
Abstract
Thyroid hormone (TH) action is mediated through two nuclear TH receptors, THRα and THRβ. Although the role of THRα is well established in bone, less is known about the relevance of THRβ-mediated signaling in bone development. On ther basis of our recent finding that TH signaling is essential for initiation and formation of secondary ossification center, we evaluated the role of THRs in mediating TH effects on epiphysial bone formation. Two-day treatment of TH-deficient Tshr(-/-) mice with TH increased THRβ1 mRNA level 3.4-fold at day 7 but had no effect on THRα1 mRNA level at the proximal tibia epiphysis. Treatment of serum-free cultures of tibias from 3-day-old mice with T3 increased THRβ1 expression 2.1- and 13-fold, respectively, at 24 and 72 h. Ten-day treatment of Tshr(-/-) newborns (days 5-14) with THRβ1 agonist GC1 at 0.2 or 2.0 μg/day increased BV/TV at day 21 by 225 and 263%, respectively, compared with vehicle treatment. Two-day treatment with GC1 (0.2 μg/day) increased expression levels of Indian hedgehog (Ihh) 100-fold, osterix 15-fold, and osteocalcin 59-fold compared with vehicle at day 7 in the proximal tibia epiphysis. Gel mobility shift assay demonstrated that a putative TH response element in the distal promoter of mouse Ihh gene interacted with THRβ1. GC1 treatment (1 nM) increased Ihh distal promoter activity 20-fold after 48 h in chondroctyes. Our data suggest a novel role for THRβ1 in secondary ossification at the epiphysis that involves transcriptional upregulation of Ihh gene.
Collapse
Affiliation(s)
- Weirong Xing
- Musculoskeletal Disease Center, Veterans Affairs Loma Linda Healthcare System, Loma Linda, California; Department of Medicine, Loma Linda University, Loma Linda, California
| | - Patrick Aghajanian
- Musculoskeletal Disease Center, Veterans Affairs Loma Linda Healthcare System, Loma Linda, California
| | - Helen Goodluck
- Musculoskeletal Disease Center, Veterans Affairs Loma Linda Healthcare System, Loma Linda, California
| | - Chandrasekhar Kesavan
- Musculoskeletal Disease Center, Veterans Affairs Loma Linda Healthcare System, Loma Linda, California; Department of Medicine, Loma Linda University, Loma Linda, California
| | - Shaohong Cheng
- Musculoskeletal Disease Center, Veterans Affairs Loma Linda Healthcare System, Loma Linda, California
| | - Sheila Pourteymoor
- Musculoskeletal Disease Center, Veterans Affairs Loma Linda Healthcare System, Loma Linda, California
| | - Heather Watt
- Musculoskeletal Disease Center, Veterans Affairs Loma Linda Healthcare System, Loma Linda, California
| | - Catrina Alarcon
- Musculoskeletal Disease Center, Veterans Affairs Loma Linda Healthcare System, Loma Linda, California
| | - Subburaman Mohan
- Musculoskeletal Disease Center, Veterans Affairs Loma Linda Healthcare System, Loma Linda, California; Department of Medicine, Loma Linda University, Loma Linda, California; Department of Orthopedics, Loma Linda University, Loma Linda, California; and Department of Biochemistry, Loma Linda University, Loma Linda, California
| |
Collapse
|
39
|
LRRK1 is critical in the regulation of B-cell responses and CARMA1-dependent NF-κB activation. Sci Rep 2016; 6:25738. [PMID: 27166870 PMCID: PMC4863158 DOI: 10.1038/srep25738] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/11/2016] [Accepted: 04/21/2016] [Indexed: 12/12/2022] Open
Abstract
B-cell receptor (BCR) signaling plays a critical role in B-cell activation and humoral immunity. In this study, we discovered a critical function of leucine-rich repeat kinase 1 (LRRK1) in BCR-mediated immune responses. Lrrk1−/− mice exhibited altered B1a-cell development and basal immunoglobulin production. In addition, these mice failed to produce IgG3 antibody in response to T cell–independent type 2 antigen due to defects in IgG3 class-switch recombination. Concomitantly, B cells lacking LRRK1 exhibited a profound defect in proliferation and survival upon BCR stimulation, which correlated with impaired BCR-mediated NF-κB activation and reduced expression of NF-κB target genes including Bcl-xL, cyclin D2, and NFATc1/αA. Furthermore, LRRK1 physically interacted and potently synergized with CARMA1 to enhance NF-κB activation. Our results reveal a critical role of LRRK1 in NF-κB signaling in B cells and the humoral immune response.
Collapse
|
40
|
Iida A, Xing W, Docx MKF, Nakashima T, Wang Z, Kimizuka M, Van Hul W, Rating D, Spranger J, Ohashi H, Miyake N, Matsumoto N, Mohan S, Nishimura G, Mortier G, Ikegawa S. Identification of biallelic LRRK1 mutations in osteosclerotic metaphyseal dysplasia and evidence for locus heterogeneity. J Med Genet 2016; 53:568-74. [PMID: 27055475 DOI: 10.1136/jmedgenet-2016-103756] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/06/2016] [Accepted: 03/19/2016] [Indexed: 01/12/2023]
Abstract
BACKGROUND Osteosclerotic metaphyseal dysplasia (OSMD) is a unique form of osteopetrosis characterised by severe osteosclerosis localised to the bone ends. The mode of inheritance is autosomal recessive. Its genetic basis is not known. OBJECTIVE To identify the disease gene for OSMD. METHODS AND RESULTS By whole exome sequencing in a boy with OSMD, we identified a homozygous 7 bp deletion (c.5938_5944delGAGTGGT) in the LRRK1 gene. His skeletal phenotype recapitulated that seen in the Lrrk1-deficient mouse. The shared skeletal hallmarks included severe sclerosis in the undermodelled metaphyses and epiphyseal margins of the tubular bones, costal ends, vertebral endplates and margins of the flat bones. The deletion is predicted to result in an elongated LRRK1 protein (p.E1980Afs*66) that lacks a part of its WD40 domains. In vitro functional studies using osteoclasts from Lrrk1-deficient mice showed that the deletion was a loss of function mutation. Genetic analysis of LRRK1 in two unrelated patients with OSMD suggested that OSMD is a genetically heterogeneous condition. CONCLUSIONS This is the first study to identify the causative gene of OSMD. Our study provides evidence that LRRK1 plays a critical role in the regulation of bone mass in humans.
Collapse
Affiliation(s)
- Aritoshi Iida
- Laboratory for Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan
| | - Weirong Xing
- Musculoskeletal Disease Center, Jerry L Pettis Memorial VA Medical Center, Loma Linda, California, USA Department of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Martine K F Docx
- Department of Paediatric Chronic Diseases and Nephrology, Queen Paola Children's Hospital, Antwerp, Belgium
| | - Tomoki Nakashima
- Department of Cell Signaling, Graduate school of Medical and Dental Science, Tokyo Medical and Dental University, Tokyo, Japan Japan Science and Technology Agency, PRESTO, Tokyo, Japan
| | - Zheng Wang
- Laboratory for Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan State Key Laboratory of Medical Molecular Biology, McKusick-Zhang Center for Genetic Medicine and Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Mamori Kimizuka
- Department of Orthopaedics, National Rehabilitation Center for Disabled Children, Tokyo, Japan
| | - Wim Van Hul
- Department of Medical Genetics, University of Antwerp and Antwerp University Hospital, Edegem, Belgium
| | - Dietz Rating
- Department of Pediatrics, St Annastiftskinderkrankenhaus, Ludwigshafen, Germany
| | - Jürgen Spranger
- Centre for Pediatrics and Adolescent Medicine, Freiburg, Germany
| | - Hirohumi Ohashi
- Division of Medical Genetics, Saitama Children's Medical Center, Saitama, Japan
| | - Noriko Miyake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Subburaman Mohan
- Musculoskeletal Disease Center, Jerry L Pettis Memorial VA Medical Center, Loma Linda, California, USA Department of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Gen Nishimura
- Department of Pediatric Imaging, Tokyo Metropolitan Children's Medical Center, Fuchu, Japan
| | - Geert Mortier
- Department of Medical Genetics, University of Antwerp and Antwerp University Hospital, Edegem, Belgium
| | - Shiro Ikegawa
- Laboratory for Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan Department of Orthopaedics, National Rehabilitation Center for Disabled Children, Tokyo, Japan
| |
Collapse
|
41
|
Wei W, Schwaid AG, Wang X, Wang X, Chen S, Chu Q, Saghatelian A, Wan Y. Ligand Activation of ERRα by Cholesterol Mediates Statin and Bisphosphonate Effects. Cell Metab 2016; 23:479-91. [PMID: 26777690 PMCID: PMC4785078 DOI: 10.1016/j.cmet.2015.12.010] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 01/23/2015] [Revised: 10/06/2015] [Accepted: 12/16/2015] [Indexed: 01/29/2023]
Abstract
Nuclear receptors (NRs) are key regulators of gene expression and physiology. Nearly half of all human NRs lack endogenous ligands including estrogen-related receptor α (ERRα). ERRα has important roles in cancer, metabolism, and skeletal homeostasis. Affinity chromatography of tissue lipidomes with the ERRα ligand-binding domain (LBD) and subsequent transcriptional assays identified cholesterol as an endogenous ERRα agonist. Perturbation of cholesterol biosynthesis or inhibition of ERRα revealed the interdependence of cholesterol and ERRα. In bone, the effects of cholesterol, statin, and bisphosphonate on osteoclastogenesis require ERRα; and consequently, cholesterol-induced bone loss or bisphosphonate osteoprotection is lost in ERRα knockout mice. Furthermore, statin induction of muscle toxicity and cholesterol suppression of macrophage cytokine secretion are impaired by loss or inhibition of ERRα. These findings reveal a key step in ERRα regulation and explain the actions of two highly prescribed drugs, statins and bisphosphonates.
Collapse
Affiliation(s)
- Wei Wei
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Adam G Schwaid
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Xueqian Wang
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xunde Wang
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shili Chen
- Clayton Foundation Laboratories of Peptide Biology and Helmsley Center for Genomic Medicine, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Qian Chu
- Clayton Foundation Laboratories of Peptide Biology and Helmsley Center for Genomic Medicine, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Alan Saghatelian
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA; Clayton Foundation Laboratories of Peptide Biology and Helmsley Center for Genomic Medicine, Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| | - Yihong Wan
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
42
|
Langston RG, Rudenko IN, Cookson MR. The function of orthologues of the human Parkinson's disease gene LRRK2 across species: implications for disease modelling in preclinical research. Biochem J 2016; 473:221-32. [PMID: 26811536 PMCID: PMC5165698 DOI: 10.1042/bj20150985] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/20/2022]
Abstract
In the period since LRRK2 (leucine-rich repeat kinase 2) was identified as a causal gene for late-onset autosomal dominant parkinsonism, a great deal of work has been aimed at understanding whether the LRRK2 protein might be a druggable target for Parkinson's disease (PD). As part of this effort, animal models have been developed to explore both the normal and the pathophysiological roles of LRRK2. However, LRRK2 is part of a wider family of proteins whose functions in different organisms remain poorly understood. In this review, we compare the information available on biochemical properties of LRRK2 homologues and orthologues from different species from invertebrates (e.g. Caenorhabditis elegans and Drosophila melanogaster) to mammals. We particularly discuss the mammalian LRRK2 homologue, LRRK1, and those species where there is only a single LRRK homologue, discussing examples where each of the LRRK family of proteins has distinct properties as well as those cases where there appear to be functional redundancy. We conclude that uncovering the function of LRRK2 orthologues will help to elucidate the key properties of human LRRK2 as well as to improve understanding of the suitability of different animal models for investigation of LRRK2-related PD.
Collapse
Affiliation(s)
- Rebekah G. Langston
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, NIA, NIH, Bethesda, MD, 20892
| | - Iakov N. Rudenko
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, NIA, NIH, Bethesda, MD, 20892
| | - Mark R. Cookson
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, NIA, NIH, Bethesda, MD, 20892
| |
Collapse
|
43
|
Wallings R, Manzoni C, Bandopadhyay R. Cellular processes associated with LRRK2 function and dysfunction. FEBS J 2015; 282:2806-26. [PMID: 25899482 PMCID: PMC4522467 DOI: 10.1111/febs.13305] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/30/2015] [Revised: 03/23/2015] [Accepted: 04/20/2015] [Indexed: 02/07/2023]
Abstract
Mutations in the leucine-rich repeat kinase 2 (LRRK2)-encoding gene are the most common cause of monogenic Parkinson's disease. The identification of LRRK2 polymorphisms associated with increased risk for sporadic Parkinson's disease, as well as the observation that LRRK2-Parkinson's disease has a pathological phenotype that is almost indistinguishable from the sporadic form of disease, suggested LRRK2 as the culprit to provide understanding for both familial and sporadic Parkinson's disease cases. LRRK2 is a large protein with both GTPase and kinase functions. Mutations segregating with Parkinson's disease reside within the enzymatic core of LRRK2, suggesting that modification of its activity impacts greatly on disease onset and progression. Although progress has been made since its discovery in 2004, there is still much to be understood regarding LRRK2's physiological and neurotoxic properties. Unsurprisingly, given the presence of multiple enzymatic domains, LRRK2 has been associated with a diverse set of cellular functions and signalling pathways including mitochondrial function, vesicle trafficking together with endocytosis, retromer complex modulation and autophagy. This review discusses the state of current knowledge on the role of LRRK2 in health and disease with discussion of potential substrates of phosphorylation and functional partners with particular emphasis on signalling mechanisms. In addition, the use of immune cells in LRRK2 research and the role of oxidative stress as a regulator of LRRK2 activity and cellular function are also discussed.
Collapse
Affiliation(s)
- Rebecca Wallings
- Reta Lila Weston Institute of Neurological Studies and Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Claudia Manzoni
- School of Pharmacy, University of Reading, UK.,UCL Institute of Neurology, London, UK
| | - Rina Bandopadhyay
- Reta Lila Weston Institute of Neurological Studies and Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| |
Collapse
|
44
|
Brommage R, Liu J, Hansen GM, Kirkpatrick LL, Potter DG, Sands AT, Zambrowicz B, Powell DR, Vogel P. High-throughput screening of mouse gene knockouts identifies established and novel skeletal phenotypes. Bone Res 2014; 2:14034. [PMID: 26273529 PMCID: PMC4472125 DOI: 10.1038/boneres.2014.34] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/16/2014] [Revised: 07/29/2014] [Accepted: 07/31/2014] [Indexed: 12/13/2022] Open
Abstract
Screening gene function in vivo is a powerful approach to discover novel drug targets. We present high-throughput screening (HTS) data for 3 762 distinct global gene knockout (KO) mouse lines with viable adult homozygous mice generated using either gene-trap or homologous recombination technologies. Bone mass was determined from DEXA scans of male and female mice at 14 weeks of age and by microCT analyses of bones from male mice at 16 weeks of age. Wild-type (WT) cagemates/littermates were examined for each gene KO. Lethality was observed in an additional 850 KO lines. Since primary HTS are susceptible to false positive findings, additional cohorts of mice from KO lines with intriguing HTS bone data were examined. Aging, ovariectomy, histomorphometry and bone strength studies were performed and possible non-skeletal phenotypes were explored. Together, these screens identified multiple genes affecting bone mass: 23 previously reported genes (Calcr, Cebpb, Crtap, Dcstamp, Dkk1, Duoxa2, Enpp1, Fgf23, Kiss1/Kiss1r, Kl (Klotho), Lrp5, Mstn, Neo1, Npr2, Ostm1, Postn, Sfrp4, Slc30a5, Slc39a13, Sost, Sumf1, Src, Wnt10b), five novel genes extensively characterized (Cldn18, Fam20c, Lrrk1, Sgpl1, Wnt16), five novel genes with preliminary characterization (Agpat2, Rassf5, Slc10a7, Slc26a7, Slc30a10) and three novel undisclosed genes coding for potential osteoporosis drug targets.
Collapse
Affiliation(s)
| | - Jeff Liu
- Lexicon Pharmaceuticals , The Woodlands, TX, USA
| | | | | | | | | | | | | | - Peter Vogel
- Lexicon Pharmaceuticals , The Woodlands, TX, USA
| |
Collapse
|
45
|
Genetic, structural, and molecular insights into the function of ras of complex proteins domains. ACTA ACUST UNITED AC 2014; 21:809-18. [PMID: 24981771 PMCID: PMC4104024 DOI: 10.1016/j.chembiol.2014.05.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/28/2014] [Revised: 05/13/2014] [Accepted: 05/28/2014] [Indexed: 11/22/2022]
Abstract
Ras of complex proteins (ROC) domains were identified in 2003 as GTP binding modules in large multidomain proteins from Dictyostelium discoideum. Research into the function of these domains exploded with their identification in a number of proteins linked to human disease, including leucine-rich repeat kinase 2 (LRRK2) and death-associated protein kinase 1 (DAPK1) in Parkinson’s disease and cancer, respectively. This surge in research has resulted in a growing body of data revealing the role that ROC domains play in regulating protein function and signaling pathways. In this review, recent advances in the structural information available for proteins containing ROC domains, along with insights into enzymatic function and the integration of ROC domains as molecular switches in a cellular and organismal context, are explored.
Collapse
|