1
|
Sobacchi C, Menale C, Crisafulli L, Ficara F. Role of RANKL Signaling in Bone Homeostasis. Physiology (Bethesda) 2025; 40:0. [PMID: 39255276 DOI: 10.1152/physiol.00031.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/19/2024] [Accepted: 09/02/2024] [Indexed: 09/12/2024] Open
Abstract
RANKL and its cognate receptor RANK are crucial regulators of bone metabolism in physiological as well as in pathological conditions. Here we go through the works that unveiled the paramount role of this signaling pathway. We focus on the RANKL cytokine, whose alterations are responsible for rare and common bone diseases. We describe recent insights on the regulation of RANKL expression, which provide new hints for the pharmacological regulation of this molecule. Based on the multiple functions exerted by RANKL (within and outside the bone tissue), we advise caution regarding the potential unintended consequences of its inhibition.
Collapse
Affiliation(s)
- Cristina Sobacchi
- Milan Unit, Institute of Genetic and Biomedical Research, National Research Council, Milan, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico Humanitas Research Hospital, Milan, Italy
| | - Ciro Menale
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," Naples, Italy
| | - Laura Crisafulli
- Milan Unit, Institute of Genetic and Biomedical Research, National Research Council, Milan, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico Humanitas Research Hospital, Milan, Italy
| | - Francesca Ficara
- Milan Unit, Institute of Genetic and Biomedical Research, National Research Council, Milan, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico Humanitas Research Hospital, Milan, Italy
| |
Collapse
|
2
|
Schiavone ML, Crisafulli L, Camisaschi C, De Simone G, Liberati FR, Palagano E, Rucci N, Ficara F, Sobacchi C. Rankl genetic deficiency and functional blockade undermine skeletal stem and progenitor cell differentiation. Stem Cell Res Ther 2024; 15:203. [PMID: 38971808 PMCID: PMC11227705 DOI: 10.1186/s13287-024-03803-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 06/16/2024] [Indexed: 07/08/2024] Open
Abstract
BACKGROUND Skeletal Stem Cells (SSCs) are required for skeletal development, homeostasis, and repair. The perspective of their wide application in regenerative medicine approaches has supported research in this field, even though so far results in the clinic have not reached expectations, possibly due also to partial knowledge of intrinsic, potentially actionable SSC regulatory factors. Among them, the pleiotropic cytokine RANKL, with essential roles also in bone biology, is a candidate deserving deep investigation. METHODS To dissect the role of the RANKL cytokine in SSC biology, we performed ex vivo characterization of SSCs and downstream progenitors (SSPCs) in mice lacking Rankl (Rankl-/-) by means of cytofluorimetric sorting and analysis of SSC populations from different skeletal compartments, gene expression analysis, and in vitro osteogenic differentiation. In addition, we assessed the effect of the pharmacological treatment with the anti-RANKL blocking antibody Denosumab (approved for therapy in patients with pathological bone loss) on the osteogenic potential of bone marrow-derived stromal cells from human healthy subjects (hBMSCs). RESULTS We found that, regardless of the ossification type of bone, osteochondral SSCs had a higher frequency and impaired differentiation along the osteochondrogenic lineage in Rankl-/- mice as compared to wild-type. Rankl-/- mice also had increased frequency of committed osteochondrogenic and adipogenic progenitor cells deriving from perivascular SSCs. These changes were not due to the peculiar bone phenotype of increased density caused by lack of osteoclast resorption (defined osteopetrosis); indeed, they were not found in another osteopetrotic mouse model, i.e., the oc/oc mouse, and were therefore not due to osteopetrosis per se. In addition, Rankl-/- SSCs and primary osteoblasts showed reduced mineralization capacity. Of note, hBMSCs treated in vitro with Denosumab had reduced osteogenic capacity compared to control cultures. CONCLUSIONS We provide for the first time the characterization of SSPCs from mouse models of severe recessive osteopetrosis. We demonstrate that Rankl genetic deficiency in murine SSCs and functional blockade in hBMSCs reduce their osteogenic potential. Therefore, we propose that RANKL is an important regulatory factor of SSC features with translational relevance.
Collapse
Affiliation(s)
- M L Schiavone
- IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, Milan, 20089, Italy
| | - L Crisafulli
- IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, Milan, 20089, Italy
- Institute for Genetic and Biomedical Research, Milan Unit, CNR, via Fantoli 16/15, Milan, 20138, Italy
| | - C Camisaschi
- Flow Cytometry Core, IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, Milan, 20089, Italy
| | - G De Simone
- Flow Cytometry Core, IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, Milan, 20089, Italy
| | - F R Liberati
- IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, Milan, 20089, Italy
| | - E Palagano
- Institute of Biosciences and Bioresources, CNR, via Madonna Del Piano 10, Sesto Fiorentino, 50019, FI, Italy
| | - N Rucci
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio - Coppito 2, L'Aquila, 67100, Italy
| | - F Ficara
- IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, Milan, 20089, Italy
- Institute for Genetic and Biomedical Research, Milan Unit, CNR, via Fantoli 16/15, Milan, 20138, Italy
| | - Cristina Sobacchi
- IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, Milan, 20089, Italy.
- Institute for Genetic and Biomedical Research, Milan Unit, CNR, via Fantoli 16/15, Milan, 20138, Italy.
| |
Collapse
|
3
|
Antonacci G, Vanna R, Ventura M, Schiavone ML, Sobacchi C, Behrouzitabar M, Polli D, Manzoni C, Cerullo G. Birefringence-induced phase delay enables Brillouin mechanical imaging in turbid media. Nat Commun 2024; 15:5202. [PMID: 38898004 PMCID: PMC11187154 DOI: 10.1038/s41467-024-49419-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024] Open
Abstract
Acoustic vibrations of matter convey fundamental viscoelastic information that can be optically retrieved by hyperfine spectral analysis of the inelastic Brillouin scattered light. Increasing evidence of the central role of the viscoelastic properties in biological processes has stimulated the rise of non-contact Brillouin microscopy, yet this method faces challenges in turbid samples due to overwhelming elastic background light. Here, we introduce a common-path Birefringence-Induced Phase Delay (BIPD) filter to disentangle the polarization states of the Brillouin and Rayleigh signals, enabling the rejection of the background light using a polarizer. We demonstrate a 65 dB extinction ratio in a single optical pass collecting Brillouin spectra in extremely scattering environments and across highly reflective interfaces. We further employ the BIPD filter to image bone tissues from a mouse model of osteopetrosis, highlighting altered biomechanical properties compared to the healthy control. Results herald new opportunities in mechanobiology where turbid biological samples remain poorly characterized.
Collapse
Affiliation(s)
| | - Renzo Vanna
- CNR-Istituto di Fotonica e Nanotecnologie, CNR-IFN, Piazza Leonardo da Vinci 32, 20133, Milano, Italy
| | - Marco Ventura
- CNR-Istituto di Fotonica e Nanotecnologie, CNR-IFN, Piazza Leonardo da Vinci 32, 20133, Milano, Italy
| | | | - Cristina Sobacchi
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089, Rozzano (Milano), Italy
- CNR-Istituto di Ricerca Genetica e Biomedica (CNR-IRGB), UOS di Milano, via Fantoli 16/15, 20138, Milano, Italy
| | - Morteza Behrouzitabar
- Dipartimento di Fisica, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133, Milano, Italy
| | - Dario Polli
- Specto Photonics, Via Giulio e Corrado Venini 18, 20127, Milano, Italy
- CNR-Istituto di Fotonica e Nanotecnologie, CNR-IFN, Piazza Leonardo da Vinci 32, 20133, Milano, Italy
- Dipartimento di Fisica, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133, Milano, Italy
| | - Cristian Manzoni
- CNR-Istituto di Fotonica e Nanotecnologie, CNR-IFN, Piazza Leonardo da Vinci 32, 20133, Milano, Italy.
| | - Giulio Cerullo
- CNR-Istituto di Fotonica e Nanotecnologie, CNR-IFN, Piazza Leonardo da Vinci 32, 20133, Milano, Italy
- Dipartimento di Fisica, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133, Milano, Italy
| |
Collapse
|
4
|
Bhadouria N, Holguin N. Osteoporosis treatments for intervertebral disc degeneration and back pain: a perspective. JBMR Plus 2024; 8:ziae048. [PMID: 38706880 PMCID: PMC11066806 DOI: 10.1093/jbmrpl/ziae048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 03/18/2024] [Accepted: 03/23/2024] [Indexed: 05/07/2024] Open
Abstract
Low back pain derived from intervertebral disc (IVD) degeneration is a debilitating spinal condition that, despite its prevalence, does not have any intermediary guidelines for pharmacological treatment between palliative care and invasive surgery. The development of treatments for the IVD is complicated by the variety of resident cell types needed to maintain the regionally distinct structural properties of the IVD that permit the safe, complex motions of the spine. Osteoporosis of the spine increases the risk of vertebral bone fracture that can increase the incidence of back pain. Fortunately, there are a variety of pharmacological treatments for osteoporosis that target osteoblasts, osteoclasts and/or osteocytes to build bone and prevent vertebral fracture. Of particular note, clinical and preclinical studies suggest that commonly prescribed osteoporosis drugs like bisphosphonates, intermittent parathyroid hormone, anti-sclerostin antibody, selective estrogen receptor modulators and anti-receptor activator of nuclear factor-kappa B ligand inhibitor denosumab may also relieve back pain. Here, we cite clinical and preclinical studies and include unpublished data to support the argument that a subset of these therapeutics for osteoporosis may alleviate low back pain by also targeting the IVD.
Collapse
Affiliation(s)
- Neharika Bhadouria
- Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47907, United States
| | - Nilsson Holguin
- Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| |
Collapse
|
5
|
Onji M, Penninger JM. RANKL and RANK in Cancer Therapy. Physiology (Bethesda) 2023; 38:0. [PMID: 36473204 DOI: 10.1152/physiol.00020.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Receptor activator of nuclear factor-κB (RANK) and its ligand (RANKL) are key regulators of mammalian physiology such as bone metabolism, immune tolerance and antitumor immunity, and mammary gland biology. Here, we explore the multiple functions of RANKL/RANK in physiology and pathophysiology and discuss underlying principles and strategies to modulate the RANKL/RANK pathway as a therapeutic target in immune-mediated cancer treatment.
Collapse
Affiliation(s)
- Masahiro Onji
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, VBC-Vienna BioCenter, Vienna, Austria
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, VBC-Vienna BioCenter, Vienna, Austria.,Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
6
|
Schulz A, Moshous D. Hematopoietic stem cell transplantation, a curative approach in infantile osteopetrosis. Bone 2023; 167:116634. [PMID: 36470372 DOI: 10.1016/j.bone.2022.116634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022]
Abstract
Most patients with osteopetrosis (OPT) can be causally and curatively treated with allogeneic hematopoietic stem cell transplantation (HSCT) because osteoclasts are derived from the HSC. However, HSCT is contraindicated in some forms of OPT, namely OPT with neurodegeneration (in all patients with OSTM1 and about half of patients with CLCN7 mutations) and OPT caused by an osteoblast defect (patients with RANKL mutations). HSCT for OPT risks serious side effects, such as transplant failure, venous occlusive disease, pulmonary hypertension, and hypercalcemic crises. Nevertheless, the success rate of HSCT has improved significantly in recent decades. This applies, in particular, to HSCT from non-HLA compatible (haploidentical) donors. Therefore, nowadays an HSCT can be discussed for intermediate OPT forms. After a successful HSCT, most patients have very good quality of life, but about two-thirds are visually impaired, and in rarer cases show motor and neurological disabilities. Early diagnosis, further improvements in transplantation procedures, and advances to improve quality-of-life after transplantation are challenges for the future.
Collapse
Affiliation(s)
- Ansgar Schulz
- Department of Paediatrics, University Medical Center Ulm, Eythstr. 24, D 89075 Ulm, Germany.
| | - Despina Moshous
- Department of Pediatric Immunology, Hematology and Rheumatology, Hôpital Universitaire Necker-Enfants Malades, AP-HP, Université Paris Cité, 149, rue de Sèvres, 75015 Paris, France.
| |
Collapse
|
7
|
Abstract
Osteopetrosis (OPT) is a rare inherited bone disease characterized by a bone resorption defect, due to osteoclast malfunction (in osteoclast-rich, oc-rich, OPT forms) or absence (in oc-poor OPT forms). This causes severe clinical abnormalities, including increased bone density, lack of bone marrow cavity, stunted growth, macrocephaly, progressive deafness, blindness, hepatosplenomegaly, and severe anemia. The oc-poor subtype of OPT is ultra-rare in humans. It is caused by mutations in either the tumor necrosis factor ligand superfamily member 11 (TNFSF11) gene, encoding RANKL (Receptor Activator of Nuclear factor-kappa B [NF-κB] Ligand) which is expressed on cells of mesenchymal origin and lymphocytes, or the TNFRSF member 11A (TNFRSF11A) gene, encoding the RANKL functional receptor RANK which is expressed on cells of myeloid lineage including osteoclasts. Clinical presentation is usually severe with onset in early infancy or in fetal life, although as more patients are reported, expressivity is variable. Phenotypic variability of RANK-deficient OPT sometimes includes hypogammaglobulinemia or radiological features of dysosteosclerosis. Disease progression is somewhat slower in RANKL-deficient OPT than in other 'malignant' subtypes of OPT. While both RANKL and RANK are essential for normal bone turnover, hematopoietic stem cell transplantation (HSCT) is the treatment of choice only for patients with the RANK-deficient form of oc-poor OPT. So far, there is no cure for RANKL-deficient OPT.
Collapse
Affiliation(s)
- Cristina Sobacchi
- CNR-IRGB, Milan Unit, via Fantoli 16/15, 20138 Milan, Italy; Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, MI, Italy.
| | - Mario Abinun
- Paediatric Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital, Royal Victoria Infirmary, Queen Victoria Road, Newcastle upon Tyne NE1 4LP, UK; Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| |
Collapse
|
8
|
Li H, Li Y, Zou J, Yang Y, Han R, Zhang J. Sinomenine Inhibits Orthodontic Tooth Movement and Root Resorption in Rats and Enhances Osteogenic Differentiation of PDLSCs. Drug Des Devel Ther 2022; 16:2949-2965. [PMID: 36090955 PMCID: PMC9462521 DOI: 10.2147/dddt.s379468] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/24/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose To investigate the effects of sinomenine on orthodontic tooth movement and root resorption in rats, as well as the effect of sinomenine on the osteogenesis of periodontal ligament stem cells (PDLSCs). Methods Fifty-four male Wistar rats were randomly divided into 3 groups: control group, 20 mg/kg sinomenine group and 40 mg/kg sinomenine group. Fifty-gram orthodontic force was applied to all groups. Each group was injected intraperitoneally with corresponding concentration of sinomenine every day. After 14 days, all rats were sacrificed. Micro-computed tomography (micro-CT) scan was used to analyze tooth movement, root resorption and alveolar bone changes. The effect on periodontal tissue was analyzed by Masson, tartrate-resistant acid phosphatase (TRAP) and immunohistochemical staining. In vitro, PDLSCs were extracted and identified. The effect of sinomenine on proliferation was determined by cell-counting kit-8. The effect of sinomenine on osteogenesis was investigated by alkaline phosphatase (ALP) activity and alizarin red staining. qPCR and Western blotting were performed to explore the effects of sinomenine on the expression levels of ALP, runt-related transcription factor 2 (RUNX2), receptor activator of nuclear factor kappaB ligand (RANKL) and osteoprotegerin (OPG). Results The tooth movement and root resorption of sinomenine groups were reduced. Sinomenine decreased trabecular spacing on compression side and increased alveolar bone volume and trabecular thickness on tension side. TRAP-positive cells in sinomenine groups decreased significantly. The expressions of TNF-α and RANKL were decreased, while the expressions of OPG, RUNX2 and osteocalcin were up-regulated. In vitro, 0.1 M and 0.5 M sinomenine enhanced ALP activity, mineral deposition and the expression of ALP, RUNX2 and OPG, and reduced the expression of RANKL. Conclusion Sinomenine could inhibit tooth movement, reduce root resorption, and exert a positive effect on bone formation in rats. Moreover, sinomenine promoted the osteogenesis of PDLSCs.
Collapse
Affiliation(s)
- Hongkun Li
- Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, People’s Republic of China
| | - Yilin Li
- Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, People’s Republic of China
| | - Jinghua Zou
- Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, People’s Republic of China
| | - Yanran Yang
- Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, People’s Republic of China
| | - Ruiqi Han
- Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, People’s Republic of China
| | - Jun Zhang
- Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, People’s Republic of China
- Correspondence: Jun Zhang, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, No. 44-1 Wenhua Road West, Jinan, 250012, People’s Republic of China, Tel +86 139 5310 9816, Email
| |
Collapse
|
9
|
Rosichini M, Catanoso M, Screpanti I, Felli MP, Locatelli F, Velardi E. Signaling Crosstalks Drive Generation and Regeneration of the Thymus. Front Immunol 2022; 13:920306. [PMID: 35734178 PMCID: PMC9207182 DOI: 10.3389/fimmu.2022.920306] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/17/2022] [Indexed: 12/19/2022] Open
Abstract
Optimal recovery of immune competence after periods of hematopoietic insults or stress is crucial to re-establish patient response to vaccines, pathogens and tumor antigens. This is particularly relevant for patients receiving high doses of chemotherapy or radiotherapy, who experience prolonged periods of lymphopenia, which can be associated with an increased risk of infections, malignant relapse, and adverse clinical outcome. While the thymus represents the primary organ responsible for the generation of a diverse pool of T cells, its function is profoundly impaired by a range of acute insults (including those caused by cytoreductive chemo/radiation therapy, infections and graft-versus-host disease) and by the chronic physiological deterioration associated with aging. Impaired thymic function increases the risk of infections and tumor antigen escape due to a restriction in T-cell receptor diversity and suboptimal immune response. Therapeutic approaches that can promote the renewal of the thymus have the potential to restore immune competence in patients. Previous work has documented the importance of the crosstalk between thymocytes and thymic epithelial cells in establishing correct architecture and function of thymic epithelium. This crosstalk is relevant not only during thymus organogenesis, but also to promote the recovery of its function after injuries. In this review, we will analyze the signals involved in the crosstalk between TECs and hematopoietic cells. We will focus in particular on how signals from T-cells can regulate TEC function and discuss the relevance of these pathways in restoring thymic function and T-cell immunity in experimental models, as well as in the clinical setting.
Collapse
Affiliation(s)
- Marco Rosichini
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Marialuigia Catanoso
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Isabella Screpanti
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Maria Pia Felli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Franco Locatelli
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- Department of Maternal and Child Health, Sapienza University of Rome, Rome, Italy
| | - Enrico Velardi
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- *Correspondence: Enrico Velardi,
| |
Collapse
|
10
|
Gulla S, Reddy MC, Reddy VC, Chitta S, Bhanoori M, Lomada D. Role of thymus in health and disease. Int Rev Immunol 2022; 42:347-363. [PMID: 35593192 DOI: 10.1080/08830185.2022.2064461] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/26/2022] [Accepted: 04/04/2022] [Indexed: 01/04/2023]
Abstract
The thymus is a primary lymphoid organ, essential for the development of T-cells that will protect from invading pathogens, immune disorders, and cancer. The thymus decreases in size and cellularity with age referred to as thymus involution or atrophy. This involution causes decreased T-cell development and decreased naive T-cell emigration to the periphery, increased proportion of memory T cells, and a restricted, altered T-cell receptor (TCR) repertoire. The changes in composition and function of the circulating T cell pool as a result of thymic involution led to increased susceptibility to infectious diseases including the recent COVID and a higher risk for autoimmune disorders and cancers. Thymic involution consisting of both structural and functional loss of the thymus has a deleterious effect on T cell development, T cell selection, and tolerance. The mechanisms which act on the structural (cortex and medulla) matrix of the thymus, the gradual accumulation of genetic mutations, and altered gene expressions may lead to immunosenescence as a result of thymus involution. Understanding the molecular mechanisms behind thymic involution is critical for identifying diagnostic biomarkers and targets for treatment help to develop strategies to mitigate thymic involution-associated complications. This review is focused on the consequences of thymic involution in infections, immune disorders, and diseases, identifying potential checkpoints and potential approaches to sustain or restore the function of the thymus particularly in elderly and immune-compromised individuals.
Collapse
Affiliation(s)
- Surendra Gulla
- Department of Biotechnology and Bioinformatics, Yogi Vemana University, Kadapa, Andhra Pradesh, India
| | - Madhava C Reddy
- Department of Biotechnology and Bioinformatics, Yogi Vemana University, Kadapa, Andhra Pradesh, India
| | - Vajra C Reddy
- Katuri Medical College and Hospital, Chinnakondrupadu, Guntur, India
| | | | - Manjula Bhanoori
- Department of Biochemistry, Osmania University, Hyderabad, Telangana State, India
| | - Dakshayani Lomada
- Department of Genetics and Genomics, Yogi Vemana University, Kadapa, Andhra Pradesh, India
| |
Collapse
|
11
|
Robinson LJ, Soboloff J, Tourkova IL, Larrouture QC, Witt MR, Gross S, Hooper R, Samakai E, Worley PF, Barnett JB, Blair HC. The function of the calcium channel Orai1 in osteoclast development. FASEB J 2021; 35:e21653. [PMID: 34009685 DOI: 10.1096/fj.202001921rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 04/22/2021] [Accepted: 04/26/2021] [Indexed: 11/11/2022]
Abstract
To determine the intrinsic role of Orai1 in osteoclast development, Orai1-floxed mice were bred with LysMcre mice to delete Orai1 from the myeloid lineage. PCR, in situ labelling and Western analysis showed Orai1 deletion in myeloid-lineage cells, including osteoclasts, as expected. Surprisingly, bone resorption was maintained in vivo, despite loss of multinucleated osteoclasts; instead, a large number of mononuclear cells bearing tartrate resistant acid phosphatase were observed on cell surfaces. An in vitro resorption assay confirmed that RANKL-treated Orai1 null cells, also TRAP-positive but mononuclear, degraded matrix, albeit at a reduced rate compared to wild type osteoclasts. This shows that mononuclear osteoclasts can degrade bone, albeit less efficiently. Further unexpected findings included that Orai1fl/fl -LysMcre vertebrae showed slightly reduced bone density in 16-week-old mice, despite Orai1 deletion only in myeloid cells; however, this mild difference resolved with age. In summary, in vitro analysis showed a severe defect in osteoclast multinucleation in Orai1 negative mononuclear cells, consistent with prior studies using less targeted strategies, but with evidence of resorption in vivo and unexpected secondary effects on bone formation leaving bone mass largely unaffected.
Collapse
Affiliation(s)
- Lisa J Robinson
- Department of Pathology, Anatomy and Laboratory Medicine, West Virginia University School of Medicine, Morgantown, WV, USA.,Department of Microbiology, Immunology & Cell Biology, West Virginia University Cancer Institute, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Jonathan Soboloff
- Fels Cancer Institute for Personalized Medicine, Department of Medical Genetics & Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Irina L Tourkova
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA.,Pittsburgh VA Medical Center, Pittsburgh, PA, USA
| | - Quitterie C Larrouture
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA.,Pittsburgh VA Medical Center, Pittsburgh, PA, USA
| | - Michelle R Witt
- Department of Pathology, Anatomy and Laboratory Medicine, West Virginia University School of Medicine, Morgantown, WV, USA.,Department of Microbiology, Immunology & Cell Biology, West Virginia University Cancer Institute, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Scott Gross
- Fels Cancer Institute for Personalized Medicine, Department of Medical Genetics & Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Robert Hooper
- Fels Cancer Institute for Personalized Medicine, Department of Medical Genetics & Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Elsie Samakai
- Fels Cancer Institute for Personalized Medicine, Department of Medical Genetics & Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Paul F Worley
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - John B Barnett
- Department of Microbiology, Immunology & Cell Biology, West Virginia University Cancer Institute, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Harry C Blair
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA.,Pittsburgh VA Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
12
|
Ferrua F, Bortolomai I, Fontana E, Di Silvestre D, Rigoni R, Marcovecchio GE, Draghici E, Brambilla F, Castiello MC, Delfanti G, Moshous D, Picard C, Taghon T, Bordon V, Schulz AS, Schuetz C, Giliani S, Soresina A, Gennery AR, Signa S, Dávila Saldaña BJ, Delmonte OM, Notarangelo LD, Roifman CM, Poliani PL, Uva P, Mauri PL, Villa A, Bosticardo M. Thymic Epithelial Cell Alterations and Defective Thymopoiesis Lead to Central and Peripheral Tolerance Perturbation in MHCII Deficiency. Front Immunol 2021; 12:669943. [PMID: 34211466 PMCID: PMC8239840 DOI: 10.3389/fimmu.2021.669943] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 05/07/2021] [Indexed: 11/13/2022] Open
Abstract
Major Histocompatibility Complex (MHC) class II (MHCII) deficiency (MHCII-D), also known as Bare Lymphocyte Syndrome (BLS), is a rare combined immunodeficiency due to mutations in genes regulating expression of MHCII molecules. MHCII deficiency results in impaired cellular and humoral immune responses, leading to severe infections and autoimmunity. Abnormal cross-talk with developing T cells due to the absence of MHCII expression likely leads to defects in thymic epithelial cells (TEC). However, the contribution of TEC alterations to the pathogenesis of this primary immunodeficiency has not been well characterized to date, in particular in regard to immune dysregulation. To this aim, we have performed an in-depth cellular and molecular characterization of TEC in this disease. We observed an overall perturbation of thymic structure and function in both MHCII-/- mice and patients. Transcriptomic and proteomic profiling of murine TEC revealed several alterations. In particular, we demonstrated that impairment of lymphostromal cross-talk in the thymus of MHCII-/- mice affects mTEC maturation and promiscuous gene expression and causes defects of central tolerance. Furthermore, we observed peripheral tolerance impairment, likely due to defective Treg cell generation and/or function and B cell tolerance breakdown. Overall, our findings reveal disease-specific TEC defects resulting in perturbation of central tolerance and limiting the potential benefits of hematopoietic stem cell transplantation in MHCII deficiency.
Collapse
Affiliation(s)
- Francesca Ferrua
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Ileana Bortolomai
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elena Fontana
- Human Genome Department, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
- Milan Unit, Institute of Genetic and Biomedical Research, National Research Council (CNR), Milan, Italy
| | - Dario Di Silvestre
- Department of Biomedical Sciences, Institute for Biomedical Technologies-National Research Council (CNR), Milan, Italy
| | - Rosita Rigoni
- Human Genome Department, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
- Milan Unit, Institute of Genetic and Biomedical Research, National Research Council (CNR), Milan, Italy
| | - Genni Enza Marcovecchio
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elena Draghici
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Brambilla
- Department of Biomedical Sciences, Institute for Biomedical Technologies-National Research Council (CNR), Milan, Italy
| | - Maria Carmina Castiello
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Milan Unit, Institute of Genetic and Biomedical Research, National Research Council (CNR), Milan, Italy
| | - Gloria Delfanti
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Despina Moshous
- Department of Pediatric Immunology, Hematology and Rheumatology, Necker Children’s Hospital, AP-HP, Paris, France
- Laboratory “Genome Dynamics in the Immune System”, INSERM UMR1163, Université de Paris, Institut Imagine, Paris, France
| | - Capucine Picard
- Department of Pediatric Immunology, Hematology and Rheumatology, Necker Children’s Hospital, AP-HP, Paris, France
- Centre d’Etude des Déficits Immunitaires, Necker-Enfants Malades Hospital, AP-HP, Paris, France
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, Inserm UMR 1163, University Paris Descartes Sorbonne Paris Cité, Imagine Institute, Paris, France
| | - Tom Taghon
- Department of Diagnostic Sciences, Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Victoria Bordon
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium
| | - Ansgar S. Schulz
- Department of Pediatrics, University Medical Center Ulm, Ulm, Germany
| | - Catharina Schuetz
- Department of Pediatrics, University Medical Center Ulm, Ulm, Germany
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Silvia Giliani
- Cytogenetics and Medical Genetics Unit and “A. Nocivelli” Institute for Molecular Medicine, Spedali Civili Hospital, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Annarosa Soresina
- Unit of Pediatric Immunology, Pediatrics Clinic, University of Brescia, ASST-Spedali Civili Brescia, Brescia, Italy
| | - Andrew R. Gennery
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Department of Pediatric Immunology and HSCT, Great North Children's Hospital, Newcastle upon Tyne, United Kingdom
| | - Sara Signa
- Department of Pediatric Immunology and HSCT, Great North Children's Hospital, Newcastle upon Tyne, United Kingdom
- Autoinflammatory Diseases and Immunodeficiencies Center, IRCCS Istituto G. Gaslini, and Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, and Maternal and Children's Sciences, University of Genoa, Genoa, Italy
| | - Blachy J. Dávila Saldaña
- Division of Blood and Marrow Transplantation, Children's National Hospital, Washington, DC, United States
| | - Ottavia M. Delmonte
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, United States
| | - Luigi D. Notarangelo
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, United States
| | - Chaim M. Roifman
- Division of Immunology & Allergy, Department of Pediatrics, The Hospital for Sick Children, the Canadian Centre for Primary Immunodeficiency and the University of Toronto, Toronto, ON, Canada
| | - Pietro Luigi Poliani
- Department of Molecular and Translational Medicine, Pathology Unit, University of Brescia, Brescia, Italy
| | - Paolo Uva
- CRS4, Science and Technology Park Polaris, Pula, Cagliari, Italy
| | - Pier Luigi Mauri
- Department of Biomedical Sciences, Institute for Biomedical Technologies-National Research Council (CNR), Milan, Italy
| | - Anna Villa
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Milan Unit, Institute of Genetic and Biomedical Research, National Research Council (CNR), Milan, Italy
| | - Marita Bosticardo
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, United States
| |
Collapse
|
13
|
Penna S, Villa A, Capo V. Autosomal recessive osteopetrosis: mechanisms and treatments. Dis Model Mech 2021; 14:261835. [PMID: 33970241 PMCID: PMC8188884 DOI: 10.1242/dmm.048940] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Autosomal recessive osteopetrosis (ARO) is a severe inherited bone disease characterized by defective osteoclast resorption or differentiation. Clinical manifestations include dense and brittle bones, anemia and progressive nerve compression, which hamper the quality of patients' lives and cause death in the first 10 years of age. This Review describes the pathogenesis of ARO and highlights the strengths and weaknesses of the current standard of care, namely hematopoietic stem cell transplantation (HSCT). Despite an improvement in the overall survival and outcomes of HSCT, transplant-related morbidity and the pre-existence of neurological symptoms significantly limit the success of HSCT, while the availability of human leukocyte antigen (HLA)-matched donors still remains an open issue. Novel therapeutic approaches are needed for ARO patients, especially for those that cannot benefit from HSCT. Here, we review preclinical and proof-of-concept studies, such as gene therapy, systematic administration of deficient protein, in utero HSCT and gene editing. Summary: Autosomal recessive osteopetrosis is a heterogeneous and rare bone disease for which effective treatments are still lacking for many patients. Here, we review the literature on clinical, preclinical and proof-of-concept studies.
Collapse
Affiliation(s)
- Sara Penna
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan 20132, Italy.,Translational and Molecular Medicine (DIMET), University of Milano-Bicocca, Monza 20900, Italy
| | - Anna Villa
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan 20132, Italy.,Institute of Genetic and Biomedical Research, Milan Unit, National Research Council, Milan 20090, Italy
| | - Valentina Capo
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan 20132, Italy.,Institute of Genetic and Biomedical Research, Milan Unit, National Research Council, Milan 20090, Italy
| |
Collapse
|
14
|
Granadier D, Iovino L, Kinsella S, Dudakov JA. Dynamics of thymus function and T cell receptor repertoire breadth in health and disease. Semin Immunopathol 2021; 43:119-134. [PMID: 33608819 PMCID: PMC7894242 DOI: 10.1007/s00281-021-00840-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/12/2021] [Indexed: 12/26/2022]
Abstract
T cell recognition of unknown antigens relies on the tremendous diversity of the T cell receptor (TCR) repertoire; generation of which can only occur in the thymus. TCR repertoire breadth is thus critical for not only coordinating the adaptive response against pathogens but also for mounting a response against malignancies. However, thymic function is exquisitely sensitive to negative stimuli, which can come in the form of acute insult, such as that caused by stress, infection, or common cancer therapies; or chronic damage such as the progressive decline in thymic function with age. Whether it be prolonged T cell deficiency after hematopoietic cell transplantation (HCT) or constriction in the breadth of the peripheral TCR repertoire with age; these insults result in poor adaptive immune responses. In this review, we will discuss the importance of thymic function for generation of the TCR repertoire and how acute and chronic thymic damage influences immune health. We will also discuss methods that are used to measure thymic function in patients and strategies that have been developed to boost thymic function.
Collapse
Affiliation(s)
- David Granadier
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA, USA
- Department of Molecular and Cellular Biology, University of Washington, Seattle, WA, USA
| | - Lorenzo Iovino
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Sinéad Kinsella
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Jarrod A Dudakov
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
- Department of Immunology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
15
|
Trajanoska K, Rivadeneira F. Genomic Medicine: Lessons Learned From Monogenic and Complex Bone Disorders. Front Endocrinol (Lausanne) 2020; 11:556610. [PMID: 33162933 PMCID: PMC7581702 DOI: 10.3389/fendo.2020.556610] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 08/21/2020] [Indexed: 12/11/2022] Open
Abstract
Current genetic studies of monogenic and complex bone diseases have broadened our understanding of disease pathophysiology, highlighting the need for medical interventions and treatments tailored to the characteristics of patients. As genomic research progresses, novel insights into the molecular mechanisms are starting to provide support to clinical decision-making; now offering ample opportunities for disease screening, diagnosis, prognosis and treatment. Drug targets holding mechanisms with genetic support are more likely to be successful. Therefore, implementing genetic information to the drug development process and a molecular redefinition of skeletal disease can help overcoming current shortcomings in pharmaceutical research, including failed attempts and appalling costs. This review summarizes the achievements of genetic studies in the bone field and their application to clinical care, illustrating the imminent advent of the genomic medicine era.
Collapse
|
16
|
Kinsella S, Dudakov JA. When the Damage Is Done: Injury and Repair in Thymus Function. Front Immunol 2020; 11:1745. [PMID: 32903477 PMCID: PMC7435010 DOI: 10.3389/fimmu.2020.01745] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 06/30/2020] [Indexed: 01/02/2023] Open
Abstract
Even though the thymus is exquisitely sensitive to acute insults like infection, shock, or common cancer therapies such as cytoreductive chemo- or radiation-therapy, it also has a remarkable capacity for repair. This phenomenon of endogenous thymic regeneration has been known for longer even than its primary function to generate T cells, however, the underlying mechanisms controlling the process have been largely unstudied. Although there is likely continual thymic involution and regeneration in response to stress and infection in otherwise healthy people, acute and profound thymic damage such as that caused by common cancer cytoreductive therapies or the conditioning regimes as part of hematopoietic cell transplantation (HCT), leads to prolonged T cell deficiency; precipitating high morbidity and mortality from opportunistic infections and may even facilitate cancer relapse. Furthermore, this capacity for regeneration declines with age as a function of thymic involution; which even at steady state leads to reduced capacity to respond to new pathogens, vaccines, and immunotherapy. Consequently, there is a real clinical need for strategies that can boost thymic function and enhance T cell immunity. One approach to the development of such therapies is to exploit the processes of endogenous thymic regeneration into novel pharmacologic strategies to boost T cell reconstitution in clinical settings of immune depletion such as HCT. In this review, we will highlight recent work that has revealed the mechanisms by which the thymus is capable of repairing itself and how this knowledge is being used to develop novel therapies to boost immune function.
Collapse
Affiliation(s)
- Sinéad Kinsella
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Jarrod A. Dudakov
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
- Department of Immunology, University of Washington, Seattle, WA, United States
| |
Collapse
|
17
|
Freese J, Greenup E, Sunil B, Ashraf AP. Osteopetrorickets Presenting with Failure to Thrive and Hypophosphatemia. J Endocr Soc 2020; 4:bvaa044. [PMID: 32500110 PMCID: PMC7255499 DOI: 10.1210/jendso/bvaa044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Indexed: 12/31/2022] Open
Abstract
Osteopetrosis is a rare group of bone disorders characterized by defective osteoclast bone resorption causing high bone mineral density. A high bone mineral density in combination with defective skeletal mineralization results in a phenotype of osteopetrorickets. We present a rare presentation of infantile osteopetrorickets in an 8-week-old female who presented with failure to thrive, hypophosphatemia, anemia, and thrombocytopenia. A skeletal survey showed increased bone density with rachitic changes. She was found to have a homozygous T-cell immune regulator 1 (TCIRG1) pathogenic mutation consistent with osteopetrosis. This highlights the importance of a clinical suspicion of osteopetrosis with this symptom constellation.
Collapse
Affiliation(s)
- Jurhee Freese
- Division of Pediatric Endocrinology and Metabolism, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Erin Greenup
- Division of Pediatric Endocrinology and Metabolism, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Bhuvana Sunil
- Division of Pediatric Endocrinology and Metabolism, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Ambika P Ashraf
- Division of Pediatric Endocrinology and Metabolism, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
18
|
Ko Y, Lee G, Kim B, Park M, Jang Y, Lim W. Modification of the RANKL-RANK-binding site for the immunotherapeutic treatment of osteoporosis. Osteoporos Int 2020; 31:983-993. [PMID: 31863125 DOI: 10.1007/s00198-019-05200-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 10/16/2019] [Indexed: 12/20/2022]
Abstract
UNLABELLED Here, we proposed the use of mutated RANKL as an immunogen for active immunization and to induce anti-cytokine antibodies for osteoporosis treatment. INTRODUCTION Osteoclasts are responsible for bone resorption in bone-related disorders. Anti-cytokine therapeutic antibodies such as denosumab are effective for the treatment of osteoporosis. However, problems with antibody manufacturing and the immunogenicity caused by multiple antibody doses have led to the use of auto-cytokines as immunogens to induce anti-cytokine antibodies. METHODS RANKL was point-mutated based on the crystal structure of the complex of RANKL and its receptor RANK. RESULTS As a proof of concept, immunization with RANKL produced high levels of specific antibodies and blocked osteoclast development in vitro and inhibited osteoporosis in RANKL-treated or ovariectomized mouse models. CONCLUSIONS The results demonstrate the successful use of mutated RANKL as an immunogen for the induction of anti-RANKL immune response. This strategy is useful in general anti-cytokine immunotherapy to avoid toxic side effects of osteoporosis treatment.
Collapse
Affiliation(s)
- Y Ko
- Department of Orthopaedic Surgery, Chosun University Hospital, Gwangju, 61453, South Korea
- Laboratory of Orthopaedic Research, Chosun University Hospital, Gwangju, 61453, South Korea
| | - G Lee
- Department of Orthopaedic Surgery, Chosun University Hospital, Gwangju, 61453, South Korea
- Laboratory of Orthopaedic Research, Chosun University Hospital, Gwangju, 61453, South Korea
| | - B Kim
- Department of Orthopaedic Surgery, Chosun University Hospital, Gwangju, 61453, South Korea
- Laboratory of Orthopaedic Research, Chosun University Hospital, Gwangju, 61453, South Korea
| | - M Park
- Department of Orthopaedic Surgery, Chosun University Hospital, Gwangju, 61453, South Korea
- Laboratory of Orthopaedic Research, Chosun University Hospital, Gwangju, 61453, South Korea
| | - Y Jang
- Department of Orthopaedic Surgery, Chosun University Hospital, Gwangju, 61453, South Korea
- Laboratory of Orthopaedic Research, Chosun University Hospital, Gwangju, 61453, South Korea
- Department of Premedical Program, School of Medicine, Chosun University, Gwangju, 61452, South Korea
| | - W Lim
- Department of Orthopaedic Surgery, Chosun University Hospital, Gwangju, 61453, South Korea.
- Laboratory of Orthopaedic Research, Chosun University Hospital, Gwangju, 61453, South Korea.
- Department of Premedical Program, School of Medicine, Chosun University, Gwangju, 61452, South Korea.
| |
Collapse
|
19
|
Generation of an immunodeficient mouse model of tcirg1-deficient autosomal recessive osteopetrosis. Bone Rep 2020; 12:100242. [PMID: 31938717 PMCID: PMC6953598 DOI: 10.1016/j.bonr.2020.100242] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/23/2019] [Accepted: 01/04/2020] [Indexed: 01/16/2023] Open
Abstract
Background Autosomal recessive osteopetrosis is a rare skeletal disorder with increased bone density due to a failure in osteoclast bone resorption. In most cases, the defect is cell-autonomous, and >50% of patients bear mutations in the TCIRG1 gene, encoding for a subunit of the vacuolar proton pump essential for osteoclast resorptive activity. The only cure is hematopoietic stem cell transplantation, which corrects the bone pathology by allowing the formation of donor-derived functional osteoclasts. Therapeutic approaches using patient-derived cells corrected ex vivo through viral transduction or gene editing can be considered, but to date functional rescue cannot be demonstrated in vivo because a relevant animal model for xenotransplant is missing. Methods We generated a new mouse model, which we named NSG oc/oc, presenting severe autosomal recessive osteopetrosis owing to the Tcirg1oc mutation, and profound immunodeficiency caused by the NSG background. We performed neonatal murine bone marrow transplantation and xenotransplantation with human CD34+ cells. Results We demonstrated that neonatal murine bone marrow transplantation rescued NSG oc/oc mice, in line with previous findings in the oc/oc parental strain and with evidence from clinical practice in humans. Importantly, we also demonstrated human cell chimerism in the bone marrow of NSG oc/oc mice transplanted with human CD34+ cells. The severity and rapid progression of the disease in the mouse model prevented amelioration of the bone pathology; nevertheless, we cannot completely exclude that minor early modifications of the bone tissue might have occurred. Conclusion Our work paves the way to generating an improved xenograft model for in vivo evaluation of functional rescue of patient-derived corrected cells. Further refinement of the newly generated mouse model will allow capitalizing on it for an optimized exploitation in the path to novel cell therapies. Ex vivo corrected autologous HSCs might cure Autosomal Recessive Osteopetrosis (ARO). There is no animal model to prove in vivo functional rescue of corrected human cells. NSG oc/oc mice display osteoclast-rich cell-autonomous ARO and immunodeficiency. Human CD34+ cell-transplanted NSG oc/oc mice show human cell chimerism in the BM. Further improvements will allow in vivo evaluating corrected patient-derived cells.
Collapse
|
20
|
Kumar G, Roger PM. From Crosstalk between Immune and Bone Cells to Bone Erosion in Infection. Int J Mol Sci 2019; 20:E5154. [PMID: 31627424 PMCID: PMC6834200 DOI: 10.3390/ijms20205154] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/05/2019] [Accepted: 10/15/2019] [Indexed: 12/31/2022] Open
Abstract
Bone infection and inflammation leads to the infiltration of immune cells at the site of infection, where they modulate the differentiation and function of osteoclasts and osteoblasts by the secretion of various cytokines and signal mediators. In recent years, there has been a tremendous effort to understand the cells involved in these interactions and the complex pathways of signal transduction and their ultimate effect on bone metabolism. These crosstalk mechanisms between the bone and immune system finally emerged, forming a new field of research called osteoimmunology. Diseases falling into the category of osteoimmunology, such as osteoporosis, periodontitis, and bone infections are considered to have a significant implication in mortality and morbidity of patients, along with affecting their quality of life. There is a much-needed research focus in this new field, as the reported data on the immunomodulation of immune cells and their signaling pathways seems to have promising therapeutic benefits for patients.
Collapse
Affiliation(s)
- Gaurav Kumar
- Unité 576, Institut National de la Santé et de la Recherche Médicale, 06200 Nice, France.
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA.
| | - Pierre-Marie Roger
- Unité 576, Institut National de la Santé et de la Recherche Médicale, 06200 Nice, France.
- Service d'Infectiologie, Hôpital Archet 1, Centre Hospitalier Universitaire de Nice, Université de Nice Sophia-Antipolis, 06200 Nice, France.
| |
Collapse
|
21
|
Sobacchi C, Menale C, Villa A. The RANKL-RANK Axis: A Bone to Thymus Round Trip. Front Immunol 2019; 10:629. [PMID: 30984193 PMCID: PMC6450200 DOI: 10.3389/fimmu.2019.00629] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 03/08/2019] [Indexed: 12/13/2022] Open
Abstract
The identification of Receptor activator of nuclear factor kappa B ligand (RANKL) and its cognate receptor Receptor activator of nuclear factor kappa B (RANK) during a search for novel tumor necrosis factor receptor (TNFR) superfamily members has dramatically changed the scenario of bone biology by providing the functional and biochemical proof that RANKL signaling via RANK is the master factor for osteoclastogenesis. In parallel, two independent studies reported the identification of mouse RANKL on activated T cells and of a ligand for osteoprotegerin on a murine bone marrow-derived stromal cell line. After these seminal findings, accumulating data indicated RANKL and RANK not only as essential players for the development and activation of osteoclasts, but also for the correct differentiation of medullary thymic epithelial cells (mTECs) that act as mediators of the central tolerance process by which self-reactive T cells are eliminated while regulatory T cells are generated. In light of the RANKL-RANK multi-task function, an antibody targeting this pathway, denosumab, is now commonly used in the therapy of bone loss diseases including chronic inflammatory bone disorders and osteolytic bone metastases; furthermore, preclinical data support the therapeutic application of denosumab in the framework of a broader spectrum of tumors. Here, we discuss advances in cellular and molecular mechanisms elicited by RANKL-RANK pathway in the bone and thymus, and the extent to which its inhibition or augmentation can be translated in the clinical arena.
Collapse
Affiliation(s)
- Cristina Sobacchi
- Milan Unit, Institute for Genetic and Biomedical Research (CNR-IRGB), Milan, Italy.,Humanitas Clinical and Research Center IRCCS, Rozzano, Italy
| | - Ciro Menale
- Milan Unit, Institute for Genetic and Biomedical Research (CNR-IRGB), Milan, Italy.,Humanitas Clinical and Research Center IRCCS, Rozzano, Italy
| | - Anna Villa
- Milan Unit, Institute for Genetic and Biomedical Research (CNR-IRGB), Milan, Italy.,San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
22
|
Penna S, Capo V, Palagano E, Sobacchi C, Villa A. One Disease, Many Genes: Implications for the Treatment of Osteopetroses. Front Endocrinol (Lausanne) 2019; 10:85. [PMID: 30837952 PMCID: PMC6389615 DOI: 10.3389/fendo.2019.00085] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 01/31/2019] [Indexed: 11/23/2022] Open
Abstract
Osteopetrosis is a condition characterized by increased bone mass due to defects in osteoclast function or formation. In the last decades, the molecular dissection of osteopetrosis has unveiled a plethora of molecular players responsible for different forms of the disease, some of which present also primary neurodegeneration that severely limits the therapy. Hematopoietic stem cell transplantation can cure the majority of them when performed in the first months of life, highlighting the relevance of an early molecular diagnosis. However, clinical management of these patients is constrained by the severity of the disease and lack of a bone marrow niche that may delay immune reconstitution. Based on osteopetrosis genetic heterogeneity and disease severity, personalized therapies are required for patients that are not candidate to bone marrow transplantation. This review briefly describes the genetics of osteopetrosis, its clinical heterogeneity, current therapy and innovative approaches undergoing preclinical evaluation.
Collapse
Affiliation(s)
- Sara Penna
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), San Raffaele Hospital, Milan, Italy
- Translational and Molecular Medicine (DIMET), University of Milano-Bicocca, Monza, Italy
| | - Valentina Capo
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), San Raffaele Hospital, Milan, Italy
| | - Eleonora Palagano
- The National Research Council (CNR) Institute for Genetic and Biomedical Research (IRGB)- CNR-IRGB, Milan Unit, Milan, Italy
- Humanitas Research Hospital, Rozzano, Italy
| | - Cristina Sobacchi
- The National Research Council (CNR) Institute for Genetic and Biomedical Research (IRGB)- CNR-IRGB, Milan Unit, Milan, Italy
- Humanitas Research Hospital, Rozzano, Italy
| | - Anna Villa
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), San Raffaele Hospital, Milan, Italy
- The National Research Council (CNR) Institute for Genetic and Biomedical Research (IRGB)- CNR-IRGB, Milan Unit, Milan, Italy
- *Correspondence: Anna Villa
| |
Collapse
|
23
|
Menale C, Campodoni E, Palagano E, Mantero S, Erreni M, Inforzato A, Fontana E, Schena F, Van't Hof R, Sandri M, Tampieri A, Villa A, Sobacchi C. Mesenchymal Stromal Cell-Seeded Biomimetic Scaffolds as a Factory of Soluble RANKL in Rankl-Deficient Osteopetrosis. Stem Cells Transl Med 2018; 8:22-34. [PMID: 30184340 PMCID: PMC6312453 DOI: 10.1002/sctm.18-0085] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 07/11/2018] [Indexed: 12/27/2022] Open
Abstract
Biomimetic scaffolds are extremely versatile in terms of chemical composition and physical properties, which can be defined to accomplish specific applications. One property that can be added is the production/release of bioactive soluble factors, either directly from the biomaterial, or from cells embedded within the biomaterial. We reasoned that pursuing this strategy would be appropriate to setup a cell‐based therapy for RANKL‐deficient autosomal recessive osteopetrosis, a very rare skeletal genetic disease in which lack of the essential osteoclastogenic factor RANKL impedes osteoclast formation. The exogenously administered RANKL cytokine is effective in achieving osteoclast formation and function in vitro and in vivo, thus, we produced murine Rankl−/− mesenchymal stromal cells (MSCs) overexpressing human soluble RANKL (hsRL) following lentiviral transduction (LVhsRL). Here, we described a three‐dimensional (3D) culture system based on a magnesium‐doped hydroxyapatite/collagen I (MgHA/Col) biocompatible scaffold closely reproducing bone physicochemical properties. MgHA/Col‐seeded murine MSCs showed improved properties, as compared to two‐dimensional (2D) culture, in terms of proliferation and hsRL production, with respect to LVhsRL‐transduced cells. When implanted subcutaneously in Rankl−/− mice, these cell constructs were well tolerated, colonized by host cells, and intensely vascularized. Of note, in the bone of Rankl−/− mice that carried scaffolds with either WT or LVhsRL‐transduced Rankl−/− MSCs, we specifically observed formation of TRAP+ cells, likely due to sRL released from the scaffolds into circulation. Thus, our strategy proved to have the potential to elicit an effect on the bone; further work is required to maximize these benefits and achieve improvements of the skeletal pathology in the treated Rankl−/− mice. Stem Cells Translational Medicine2019;8:22–34
Collapse
Affiliation(s)
- Ciro Menale
- CNR-IRGB, Milan Unit, Milan, Italy.,Humanitas Clinical and Research Institute, Rozzano, Italy
| | | | - Eleonora Palagano
- Humanitas Clinical and Research Institute, Rozzano, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Stefano Mantero
- CNR-IRGB, Milan Unit, Milan, Italy.,Humanitas Clinical and Research Institute, Rozzano, Italy
| | - Marco Erreni
- Humanitas Clinical and Research Institute, Rozzano, Italy
| | - Antonio Inforzato
- Humanitas Clinical and Research Institute, Rozzano, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Elena Fontana
- CNR-IRGB, Milan Unit, Milan, Italy.,Humanitas Clinical and Research Institute, Rozzano, Italy
| | - Francesca Schena
- Clinica Pediatrica e Reumatologia, UOSD Centro Malattie Autoinfiammatorie e Immunodeficienze, Genoa, Italy
| | - Rob Van't Hof
- Bone Research Group, Institute of Ageing & Chronic Disease, University of Liverpool, Liverpool, UK
| | | | | | - Anna Villa
- CNR-IRGB, Milan Unit, Milan, Italy.,Humanitas Clinical and Research Institute, Rozzano, Italy
| | - Cristina Sobacchi
- CNR-IRGB, Milan Unit, Milan, Italy.,Humanitas Clinical and Research Institute, Rozzano, Italy
| |
Collapse
|
24
|
Lee G, Ko Y, Park M, Kim B, Hyun H, Lim W. Recombinant DNA cloning of the active region of the receptor activator of NF-κB ligand (RANKL) gene and its role in osteoclastogenesis. BIOTECHNOL BIOPROC E 2018. [DOI: 10.1007/s12257-017-0279-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
25
|
Cappariello A, Loftus A, Muraca M, Maurizi A, Rucci N, Teti A. Osteoblast-Derived Extracellular Vesicles Are Biological Tools for the Delivery of Active Molecules to Bone. J Bone Miner Res 2018; 33:517-533. [PMID: 29091316 DOI: 10.1002/jbmr.3332] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 10/21/2017] [Accepted: 10/31/2017] [Indexed: 12/31/2022]
Abstract
Extracellular vesicles (EVs) are newly appreciated regulators of tissue homeostasis and a means of intercellular communication. Reports have investigated the role of EVs and their cargoes in cellular regulation and have tried to fine-tune their biotechnological use, but to date very little is known on their function in bone biology. To investigate the relevance of EV-mediated communication between bone cells, we isolated EVs from primary mouse osteoblasts and assessed membrane integrity, size, and structure by transmission electron microscopy (TEM) and fluorescence-activated cell sorting (FACS). EVs actively shuttled loaded fluorochromes to osteoblasts, monocytes, and endothelial cells. Moreover, osteoblast EVs contained mRNAs shared with donor cells. Osteoblasts are known to regulate osteoclastogenesis, osteoclast survival, and osteoclast function by the pro-osteoclastic cytokine, receptor activator of nuclear factor κ-B ligand (Rankl). Osteoblast EVs were enriched in Rankl, which increased after PTH treatment. These EVs were biologically active, supporting osteoclast survival. EVs isolated from rankl-/- osteoblasts lost this pro-osteoclastic function, indicating its Rankl-dependence. They integrated ex vivo into murine calvariae, and EV-shuttled fluorochromes were quickly taken up by the bone upon in vivo EV systemic administration. Rankl-/- mice lack the osteoclast lineage and are negative for its specific marker tartrate-resistant acid phosphatase (TRAcP). Treatment of rankl-/- mice with wild-type osteoblast EVs induced the appearance of TRAcP-positive cells in an EV density-dependent manner. Finally, osteoblast EVs internalized and shuttled anti-osteoclast drugs (zoledronate and dasatinib), inhibiting osteoclast activity in vitro and in vivo. We conclude that osteoblast EVs are involved in intercellular communication between bone cells, contribute to the Rankl pro-osteoclastic effect, and shuttle anti-osteoclast drugs, representing a potential means of targeted therapeutic delivery. © 2017 The Authors. Journal of Bone and Mineral Research Published by Wiley Periodicals Inc.
Collapse
Affiliation(s)
- Alfredo Cappariello
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Alexander Loftus
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Maurizio Muraca
- Department of Women's and Children's Health, University of Padua, Padua, Italy
| | - Antonio Maurizi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Nadia Rucci
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Anna Teti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
26
|
Bacon S, Crowley R. Developments in rare bone diseases and mineral disorders. Ther Adv Chronic Dis 2018; 9:51-60. [PMID: 29344330 PMCID: PMC5761943 DOI: 10.1177/2040622317739538] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 10/04/2017] [Indexed: 12/16/2022] Open
Abstract
In the last decade, there have been a number of significant advances made in the field of rare bone diseases. In this review, we discuss the expansion of the classification system for osteogenesis imperfecta (OI) and the resultant increase in therapeutic options available for management of OI. Bisphosphonates remain the most widely used intervention for OI, although the effect on fracture rate reduction is equivocal. We review the other therapies showing promising results, including denosumab, teriparatide, sclerostin, transforming growth factor β inhibition and gene targeted approaches. X-linked hypophosphataemia (XLH) is the most common heritable form of osteomalacia and rickets caused by a mutation in the phosphate regulating endopeptidase gene resulting in elevated serum fibroblast growth factor 23 (FGF23) and decreased renal phosphate reabsorption. The traditional treatment is phosphate replacement. We discuss the development of a human anti-FGF23 antibody (KRN23) as a promising development in the treatment of XLH. The current management of primary hypoparathyroidism is replacement with calcium and active vitamin D. This can be associated with under or over replacement and its inherent complications. We review the use of recombinant parathyroid hormone (1-84), which can significantly reduce the requirements for calcium and vitamin D resulting in greater safety and quality of life for individuals with hypoparathyroidism. The use of receptor activator of nuclear factor κB ligand infusions in the treatment of a particular form of osteopetrosis and enzyme replacement therapy for hypophosphatasia are also discussed.
Collapse
|
27
|
Woodcock TM, Frugier T, Nguyen TT, Semple BD, Bye N, Massara M, Savino B, Besio R, Sobacchi C, Locati M, Morganti-Kossmann MC. The scavenging chemokine receptor ACKR2 has a significant impact on acute mortality rate and early lesion development after traumatic brain injury. PLoS One 2017; 12:e0188305. [PMID: 29176798 PMCID: PMC5703564 DOI: 10.1371/journal.pone.0188305] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 11/04/2017] [Indexed: 12/17/2022] Open
Abstract
The atypical chemokine receptor ACKR2 promotes resolution of acute inflammation by operating as a scavenger receptor for inflammatory CC chemokines in several experimental models of inflammatory disorders, however its role in the brain remains unclear. Based on our previous reports of increased expression of inflammatory chemokines and their corresponding receptors following traumatic brain injury (TBI), we hypothesised that ACKR2 modulates neuroinflammation following brain trauma and that its deletion exacerbates cellular inflammation and chemokine production. We demonstrate increased CCL2 and ACKR2 mRNA expression in post-mortem human brain, whereby ACKR2 mRNA levels correlated with later times post-TBI. This data is consistent with the transient upregulation of ACKR2 observed in mouse brain after closed head injury (CHI). As compared to WT animals, ACKR2-/- mice showed a higher mortality rate after CHI, while the neurological outcome in surviving mice was similar. At day 1 post-injury, ACKR2-/- mice displayed aggravated lesion volume and no differences in CCL2 expression and macrophage recruitment relative to WT mice. Reciprocal regulation of ACKR2 and CCL2 expression was explored in cultured astrocytes, which are recognized as the major source of CCL2 and also express ACKR2. ACKR2 mRNA increased as early as 2 hours after an inflammatory challenge in WT astrocytes. As expected, CCL2 expression also dramatically increased at 4 hours in WT astrocytes but was significantly lower in ACKR2-/- astrocytes, possibly indicating a co-regulation of CCL2 and ACKR2 in these cells. Conversely, in vivo, CCL2 mRNA/protein levels were increased similarly in ACKR2-/- and WT brains at 4 and 12 hours after CHI, in line with the lack of differences in cerebral macrophage recruitment and neurological recovery. In conclusion, ACKR2 is induced after TBI and has a significant impact on mortality and lesion development acutely following CHI, while its role in chemokine expression, macrophage activation, brain pathology, and neurological recovery at later time-points is minor. Concordant to evidence in multiple sclerosis experimental models, our data corroborate a distinct role for ACKR2 in cerebral inflammatory processes compared to its reported functions in peripheral tissues.
Collapse
MESH Headings
- Animals
- Astrocytes/metabolism
- Astrocytes/pathology
- Bone and Bones/pathology
- Brain/metabolism
- Brain/pathology
- Brain/physiopathology
- Brain Injuries, Traumatic/genetics
- Brain Injuries, Traumatic/metabolism
- Brain Injuries, Traumatic/mortality
- Brain Injuries, Traumatic/physiopathology
- Cells, Cultured
- Chemokine CCL2/genetics
- Chemokine CCL2/metabolism
- Gene Deletion
- Humans
- Inflammation/pathology
- Macrophages/metabolism
- Macrophages/pathology
- Male
- Mice, Inbred C57BL
- Mortality
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Chemokine/genetics
- Receptors, Chemokine/metabolism
- Recovery of Function
- Up-Regulation/genetics
Collapse
Affiliation(s)
- Thomas M. Woodcock
- National Trauma Research Institute, The Alfred Hospital, Melbourne, Australia
- Department of Surgery, Monash University, Melbourne, Australia
| | - Tony Frugier
- Department of Pharmacology and Therapeutics School of Biomedical Sciences, The University of Melbourne, Melboune, Australia
| | - Tan Thanh Nguyen
- National Trauma Research Institute, The Alfred Hospital, Melbourne, Australia
- Department of Surgery, Monash University, Melbourne, Australia
| | - Bridgette Deanne Semple
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Melbourne, Australia
| | - Nicole Bye
- Division of Pharmacy, School of Medicine, University of Tasmania, Hobart, Australia
| | - Matteo Massara
- Humanitas Clinical and Research Center, Rozzano, Italy
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Benedetta Savino
- Humanitas Clinical and Research Center, Rozzano, Italy
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Roberta Besio
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Cristina Sobacchi
- Humanitas Clinical and Research Center, Rozzano, Italy
- Istituto di Ricerca Genetica e Biomedica Milan Unit, National Research Council, Milan, Italy
| | - Massimo Locati
- Humanitas Clinical and Research Center, Rozzano, Italy
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
- * E-mail: (MCMK); (ML)
| | - Maria Cristina Morganti-Kossmann
- Department of Epidemiology and Preventive Medicine, and Australian New Zealand Intensive Care Research Centre, Monash University, Melbourne, Australia
- Barrow Neurological Institute, Department of Child Health, University of Arizona, Phoenix, AZ, United States of America
- * E-mail: (MCMK); (ML)
| |
Collapse
|
28
|
Teti A, Econs MJ. Osteopetroses, emphasizing potential approaches to treatment. Bone 2017; 102:50-59. [PMID: 28167345 DOI: 10.1016/j.bone.2017.02.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 02/02/2017] [Accepted: 02/02/2017] [Indexed: 12/22/2022]
Abstract
Osteopetroses are a heterogeneous group of rare genetic bone diseases sharing the common hallmarks of reduced osteoclast activity, increased bone mass and high bone fragility. Osteoclasts are bone resorbing cells that contribute to bone growth and renewal through the erosion of the mineralized matrix. Alongside the bone forming activity by osteoblasts, osteoclasts allow the skeleton to grow harmonically and maintain a healthy balance between bone resorption and formation. Osteoclast impairment in osteopetroses prevents bone renewal and deteriorates bone quality, causing atraumatic fractures. Osteopetroses vary in severity and are caused by mutations in a variety of genes involved in bone resorption or in osteoclastogenesis. Frequent signs and symptoms include osteosclerosis, deformity, dwarfism and narrowing of the bony canals, including the nerve foramina, leading to hematological and neural failures. The disease is autosomal, with only one extremely rare form associated so far to the X-chromosome, and can have either recessive or dominant inheritance. Recessive ostepetroses are generally lethal in infancy or childhood, with a few milder forms clinically denominated intermediate osteopetroses. Dominant osteopetrosis is so far associated only with mutations in the CLCN7 gene and, although described as a benign form, it can be severely debilitating, although not at the same level as recessive forms, and can rarely result in reduced life expectancy. Severe osteopetroses due to osteoclast autonomous defects can be treated by Hematopoietic Stem Cell Transplant (HSCT), but those due to deficiency of the pro-osteoclastogenic cytokine, RANKL, are not suitable for this procedure. Likewise, it is unclear as to whether HSCT, which has high intrinsic risks, results in clinical improvement in autosomal dominant osteopetrosis. Therefore, there is an unmet medical need to identify new therapies and studies are currently in progress to test gene and cell therapies, small interfering RNA approach and novel pharmacologic treatments.
Collapse
Affiliation(s)
- Anna Teti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, via Vetoio-Coppito 2, 67100 L'Aquila, Italy.
| | - Michael J Econs
- Department of Medicine, Indiana University, 1120 W. Michigan St., Indianapolis, IN 46202, USA; Department of Medical and Molecular Genetics, Indiana University, 1120 W. Michigan St., Indianapolis, IN 46202, USA.
| |
Collapse
|
29
|
Schena F, Menale C, Caci E, Diomede L, Palagano E, Recordati C, Sandri M, Tampieri A, Bortolomai I, Capo V, Pastorino C, Bertoni A, Gattorno M, Martini A, Villa A, Traggiai E, Sobacchi C. Murine Rankl -/- Mesenchymal Stromal Cells Display an Osteogenic Differentiation Defect Improved by a RANKL-Expressing Lentiviral Vector. Stem Cells 2017; 35:1365-1377. [PMID: 28100034 DOI: 10.1002/stem.2574] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 12/02/2016] [Accepted: 12/26/2016] [Indexed: 01/08/2023]
Abstract
Autosomal recessive osteopetrosis (ARO) is a severe bone disease characterized by increased bone density due to impairment in osteoclast resorptive function or differentiation. Hematopoietic stem cell transplantation is the only available treatment; however, this therapy is not effective in RANKL-dependent ARO, since in bone this gene is mainly expressed by cells of mesenchymal origin. Of note, whether lack of RANKL production might cause a defect also in the bone marrow (BM) stromal compartment, possibly contributing to the pathology, is unknown. To verify this possibility, we generated and characterized BM mesenchymal stromal cell (BM-MSC) lines from wild type and Rankl-/- mice, and found that Rankl-/- BM-MSCs displayed reduced clonogenicity and osteogenic capacity. The differentiation defect was significantly improved by lentiviral transduction of Rankl-/- BM-MSCs with a vector stably expressing human soluble RANKL (hsRANKL). Expression of Rankl receptor, Rank, on the cytoplasmic membrane of BM-MSCs pointed to the existence of an autocrine loop possibly activated by the secreted cytokine. Based on the close resemblance of RANKL-defective osteopetrosis in humans and mice, we expect that our results are also relevant for RANKL-dependent ARO patients. Data obtained in vitro after transduction with a lentiviral vector expressing hsRANKL would suggest that restoration of RANKL production might not only rescue the defective osteoclastogenesis of this ARO form, but also improve a less obvious defect in the osteoblast lineage, thus possibly achieving higher benefit for the patients, when the approach is translated to clinics. Stem Cells 2017;35:1365-1377.
Collapse
Affiliation(s)
- Francesca Schena
- Laboratory of Immunology and Rheumatic Diseases, Pediatrics II, Genova, Italy
| | - Ciro Menale
- Milan Unit, CNR-IRGB, Milan, Italy.,Laboratory of Human Genome, Humanitas Clinical and Research Institute, Rozzano, Italy
| | - Emanuela Caci
- Molecular Genetics Laboratory G. Gaslini Children's Hospital, Genova, Italy
| | - Lorenzo Diomede
- Milan Unit, CNR-IRGB, Milan, Italy.,Laboratory of Human Genome, Humanitas Clinical and Research Institute, Rozzano, Italy
| | - Eleonora Palagano
- Laboratory of Human Genome, Humanitas Clinical and Research Institute, Rozzano, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Camilla Recordati
- Mouse and Animal Pathology Laboratory, Fondazione Filarete, Milano, Italy
| | - Monica Sandri
- ISTEC-CNR, Institute of Science & Technology for Ceramics, National Research Council of Italy, Faenza, Italy
| | - Anna Tampieri
- ISTEC-CNR, Institute of Science & Technology for Ceramics, National Research Council of Italy, Faenza, Italy
| | - Ileana Bortolomai
- Milan Unit, CNR-IRGB, Milan, Italy.,San Raffaele Telethon Institute for Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | - Valentina Capo
- San Raffaele Telethon Institute for Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | - Claudia Pastorino
- Laboratory of Immunology and Rheumatic Diseases, Pediatrics II, Genova, Italy
| | - Arinna Bertoni
- Laboratory of Immunology and Rheumatic Diseases, Pediatrics II, Genova, Italy.,Centre of Excellence for Biomedical Research, University of Genoa, Genova, Italy
| | - Marco Gattorno
- Laboratory of Immunology and Rheumatic Diseases, Pediatrics II, Genova, Italy
| | - Alberto Martini
- Laboratory of Immunology and Rheumatic Diseases, Pediatrics II, Genova, Italy
| | - Anna Villa
- Milan Unit, CNR-IRGB, Milan, Italy.,Laboratory of Human Genome, Humanitas Clinical and Research Institute, Rozzano, Italy
| | - Elisabetta Traggiai
- Laboratory of Immunology and Rheumatic Diseases, Pediatrics II, Genova, Italy.,Novartis Institute for Biomedical Research, Klybeckstrasse, Basel, Switzerland
| | - Cristina Sobacchi
- Milan Unit, CNR-IRGB, Milan, Italy.,Laboratory of Human Genome, Humanitas Clinical and Research Institute, Rozzano, Italy
| |
Collapse
|
30
|
van Delft MAM, Huitema LFA, Tas SW. The contribution of NF-κB signalling to immune regulation and tolerance. Eur J Clin Invest 2015; 45:529-39. [PMID: 25735405 DOI: 10.1111/eci.12430] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 03/02/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND Immune regulation is necessary to control inflammatory responses and to prevent autoimmune diseases. Therefore, mechanisms of central and peripheral tolerance have evolved to ensure that T cells recognize antigens as self- or non-self-antigens. The thymus is crucially important for central tolerance induction to self-antigens via negative selection of T cells. However, if T cells escape negative selection in the thymus and enter the periphery, peripheral mechanisms are active to warrant immune tolerance. Secondary lymphoid organs, as well as tolerogenic dendritic cells and regulatory T cells, play an important role in peripheral tolerance. In chronic inflammatory diseases, tertiary lymphoid organs are sometimes formed that may also be involved in the induction of peripheral tolerance. This review discusses the main processes that are involved in immune regulation and tolerance, and focuses on the contribution of NF-κB signalling to these processes. MATERIAL AND METHODS This narrative review is based on peer-reviewed publications listed on PubMed up to December 2014. The focus of our literature search was on studies investigating the role of (non)canonical NF-κB signalling in central and peripheral mechanisms of tolerance. Only studies published in English language were considered. RESULTS This review discusses the immune phenotype of mutant mice with defective (non)canonical NF-κB signalling, corroborated with human data, and emphasizes the contribution of the noncanonical NF-κB pathway to immune regulation and tolerance induction. CONCLUSIONS Noncanonical NF-κB signalling has an important immunoregulatory role in the immune system and contributes to both central and peripheral mechanisms of tolerance.
Collapse
Affiliation(s)
- Myrthe A M van Delft
- Amsterdam Rheumatology & immunology Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | | |
Collapse
|
31
|
Marrella V, Lo Iacono N, Fontana E, Sobacchi C, Sic H, Schena F, Sereni L, Castiello MC, Poliani PL, Vezzoni P, Cassani B, Traggiai E, Villa A. IL-10 Critically Modulates B Cell Responsiveness in Rankl−/− Mice. THE JOURNAL OF IMMUNOLOGY 2015; 194:4144-53. [DOI: 10.4049/jimmunol.1401977] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 02/23/2015] [Indexed: 11/19/2022]
|
32
|
Cappariello A, Paone R, Maurizi A, Capulli M, Rucci N, Muraca M, Teti A. Biotechnological approach for systemic delivery of membrane Receptor Activator of NF-κB Ligand (RANKL) active domain into the circulation. Biomaterials 2015; 46:58-69. [PMID: 25678116 PMCID: PMC4337851 DOI: 10.1016/j.biomaterials.2014.12.033] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 12/05/2014] [Accepted: 12/20/2014] [Indexed: 01/01/2023]
Abstract
Deficiency of Receptor Activator of NF-κB Ligand (RANKL) prevents osteoclast formation causing osteopetrosis. RANKL is a membrane-bound protein cleaved into active soluble (s)RANKL by metalloproteinase 14 (MMP14). We created a bio-device that harbors primary osteoblasts, cultured on 3D hydroxyapatite scaffolds carrying immobilized MMP14 catalytic domain. Scaffolds were sealed in diffusion chambers and implanted in RANKL-deficient mice. Mice received 1 or 2 diffusion chambers, once or twice and were sacrificed after 1 or 2 months from implants. A progressive increase of body weight was observed in the implanted groups. Histological sections of tibias of non-implanted mice were negative for the osteoclast marker Tartrate-Resistant Acid Phosphatase (TRAcP), consistent with the lack of osteoclasts. In contrast, tibias excised from implanted mice showed TRAcP-positive cells in the bone marrow and on the bone surface, these latter morphologically similar to mature osteoclasts. In mice implanted with 4 diffusion chambers total, we noted the highest number and size of TRAcP-positive cells, with quantifiable eroded bone surface and significant reduction of trabecular bone volume. These data demonstrate that our bio-device delivers effective sRANKL, inducing osteoclastogenesis in RANKL-deficient mice, supporting the feasibility of an innovative experimental strategy to treat systemic cytokine deficiencies.
Collapse
Affiliation(s)
- Alfredo Cappariello
- Regenerative Medicine Unit, Ospedale Pediatrico Bambino Gesù Istituto di Ricovero e Cura a Carattere Scientifico, Piazza S. Onofrio 4, 00165 Rome, Italy
| | - Riccardo Paone
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio - Coppito 2, 67100 L'Aquila, Italy
| | - Antonio Maurizi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio - Coppito 2, 67100 L'Aquila, Italy
| | - Mattia Capulli
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio - Coppito 2, 67100 L'Aquila, Italy
| | - Nadia Rucci
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio - Coppito 2, 67100 L'Aquila, Italy
| | - Maurizio Muraca
- Regenerative Medicine Unit, Ospedale Pediatrico Bambino Gesù Istituto di Ricovero e Cura a Carattere Scientifico, Piazza S. Onofrio 4, 00165 Rome, Italy
| | - Anna Teti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio - Coppito 2, 67100 L'Aquila, Italy.
| |
Collapse
|
33
|
Maxhimer JB, Bradley JP, Lee JC. Signaling pathways in osteogenesis and osteoclastogenesis: Lessons from cranial sutures and applications to regenerative medicine. Genes Dis 2015; 2:57-68. [PMID: 25961069 PMCID: PMC4425620 DOI: 10.1016/j.gendis.2014.12.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
One of the simplest models for examining the interplay between bone formation and resorption is the junction between the cranial bones. Although only roughly a quarter of patients diagnosed with craniosynostosis have been linked to known genetic disturbances, the molecular mechanisms elucidated from these studies have provided basic knowledge of bone homeostasis. This work has translated to methods and advances in bone tissue engineering. In this review, we examine the current knowledge of cranial suture biology derived from human craniosynostosis syndromes and discuss its application to regenerative medicine.
Collapse
Affiliation(s)
- Justin B. Maxhimer
- Division of Plastic and Reconstructive Surgery, UCLA David Geffen School of Medicine, CA, USA
| | - James P. Bradley
- Division of Plastic and Reconstructive Surgery, Temple University/St. Christopher's Hospital for Children, PA, USA
| | - Justine C. Lee
- Division of Plastic and Reconstructive Surgery, UCLA David Geffen School of Medicine, CA, USA
- Division of Plastic and Reconstructive Surgery, Greater Los Angeles VA Healthcare System, USA
- Corresponding author. UCLA Division of Plastic and Reconstructive Surgery, 200 UCLA Medical Plaza, Suite 465, Los Angeles, CA 90095, USA. Tel.: +1 310 794 7616; fax: +1 310 206 6833.
| |
Collapse
|
34
|
Coudert AE, de Vernejoul MC, Muraca M, Del Fattore A. Osteopetrosis and its relevance for the discovery of new functions associated with the skeleton. Int J Endocrinol 2015; 2015:372156. [PMID: 25873953 PMCID: PMC4385565 DOI: 10.1155/2015/372156] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 10/16/2014] [Accepted: 10/30/2014] [Indexed: 01/29/2023] Open
Abstract
Osteopetrosis is a rare genetic disorder characterized by an increase of bone mass due to defective osteoclast function. Patients typically displayed spontaneous fractures, anemia, and in the most severe forms hepatosplenomegaly and compression of cranial facial nerves leading to deafness and blindness. Osteopetrosis comprises a heterogeneous group of diseases as several forms are known with different models of inheritance and severity from asymptomatic to lethal. This review summarizes the genetic and clinical features of osteopetrosis, emphasizing how recent studies of this disease have contributed to understanding the central role of the skeleton in the whole body physiology. In particular, the interplay of bone with the stomach, insulin metabolism, male fertility, the immune system, bone marrow, and fat is described.
Collapse
Affiliation(s)
- Amélie E. Coudert
- Institut National de la Santé et de la Recherche Médicale U1138, Centre de Recherche des Cordeliers, Paris, France
| | | | - Maurizio Muraca
- Regenerative Medicine Unit, Bambino Gesù Children's Hospital, IRCCS, Piazza Sant'Onofrio 4, 00165 Rome, Italy
| | - Andrea Del Fattore
- Regenerative Medicine Unit, Bambino Gesù Children's Hospital, IRCCS, Piazza Sant'Onofrio 4, 00165 Rome, Italy
- *Andrea Del Fattore:
| |
Collapse
|
35
|
Rinotas V, Niti A, Dacquin R, Bonnet N, Stolina M, Han CY, Kostenuik P, Jurdic P, Ferrari S, Douni E. Novel genetic models of osteoporosis by overexpression of human RANKL in transgenic mice. J Bone Miner Res 2014; 29:1158-69. [PMID: 24127173 DOI: 10.1002/jbmr.2112] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 09/10/2013] [Accepted: 09/30/2013] [Indexed: 11/10/2022]
Abstract
Receptor activator of NF-κB ligand (RANKL) plays a key role in osteoclast-induced bone resorption across a range of degenerative bone diseases, and its specific inhibition has been recently approved as a treatment for women with postmenopausal osteoporosis at high or increased risk of fracture in the United States and globally. In the present study, we generated transgenic mice (TghuRANKL) carrying the human RANKL (huRANKL) genomic region and achieved a physiologically relevant pattern of RANKL overexpression in order to establish novel genetic models for assessing skeletal and extraskeletal pathologies associated with excessive RANKL and for testing clinical therapeutic candidates that inhibit human RANKL. TghuRANKL mice of both sexes developed early-onset bone loss, and the levels of huRANKL expression were correlated with bone resorption and disease severity. Low copy Tg5516 mice expressing huRANKL at low levels displayed a mild osteoporotic phenotype as shown by trabecular bone loss and reduced biomechanical properties. Notably, overexpression of huRANKL, in the medium copy Tg5519 line, resulted in severe early-onset osteoporosis characterized by lack of trabecular bone, destruction of the growth plate, increased osteoclastogenesis, bone marrow adiposity, increased bone remodeling, and severe cortical bone porosity accompanied by decreased bone strength. An even more severe skeletal phenotype developed in the high copy Tg5520 founder with extensive soft tissue calcification. Model validation was further established by evidence that denosumab, an antibody that inhibits human but not murine RANKL, fully corrected the hyper-resorptive and osteoporotic phenotypes of Tg5519 mice. Furthermore, overexpression of huRANKL rescued osteopetrotic phenotypes of RANKL-defective mice. These novel huRANKL transgenic models of osteoporosis represent an important advance for understanding the pathogenesis and treatment of high-turnover bone diseases and other disease states caused by excessive RANKL.
Collapse
Affiliation(s)
- Vagelis Rinotas
- Laboratory of Genetics, Department of Biotechnology, Agricultural University of Athens, Athens, Greece; Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Skeletal dysplasias with increased bone density: Evolution of molecular pathogenesis in the last century. Gene 2013; 528:41-5. [DOI: 10.1016/j.gene.2013.04.069] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2012] [Revised: 04/17/2013] [Accepted: 04/18/2013] [Indexed: 02/07/2023]
|
37
|
Sobacchi C, Schulz A, Coxon FP, Villa A, Helfrich MH. Osteopetrosis: genetics, treatment and new insights into osteoclast function. Nat Rev Endocrinol 2013; 9:522-36. [PMID: 23877423 DOI: 10.1038/nrendo.2013.137] [Citation(s) in RCA: 388] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Osteopetrosis is a genetic condition of increased bone mass, which is caused by defects in osteoclast formation and function. Both autosomal recessive and autosomal dominant forms exist, but this Review focuses on autosomal recessive osteopetrosis (ARO), also known as malignant infantile osteopetrosis. The genetic basis of this disease is now largely uncovered: mutations in TCIRG1, CLCN7, OSTM1, SNX10 and PLEKHM1 lead to osteoclast-rich ARO (in which osteoclasts are abundant but have severely impaired resorptive function), whereas mutations in TNFSF11 and TNFRSF11A lead to osteoclast-poor ARO. In osteoclast-rich ARO, impaired endosomal and lysosomal vesicle trafficking results in defective osteoclast ruffled-border formation and, hence, the inability to resorb bone and mineralized cartilage. ARO presents soon after birth and can be fatal if left untreated. However, the disease is heterogeneous in clinical presentation and often misdiagnosed. This article describes the genetics of ARO and discusses the diagnostic role of next-generation sequencing methods. The management of affected patients, including guidelines for the indication of haematopoietic stem cell transplantation (which can provide a cure for many types of ARO), are outlined. Finally, novel treatments, including preclinical data on in utero stem cell treatment, RANKL replacement therapy and denosumab therapy for hypercalcaemia are also discussed.
Collapse
Affiliation(s)
- Cristina Sobacchi
- Unit Of Support/Institute of Genetic and Biomedical Research, Milan Unit, National Research Council, Humanitas Clinical and Research Centre, Via Manzoni 113, 20089 Rozzano, Italy
| | | | | | | | | |
Collapse
|
38
|
Bollerslev J, Henriksen K, Nielsen MF, Brixen K, Van Hul W. Autosomal dominant osteopetrosis revisited: lessons from recent studies. Eur J Endocrinol 2013; 169:R39-57. [PMID: 23744590 DOI: 10.1530/eje-13-0136] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Systematic studies of autosomal dominant osteopetrosis (ADO) were followed by the identification of underlying mutations giving unique possibilities to perform translational studies. What was previously designated ADO1 turned out to be a high bone mass phenotype caused by a missense mutation in the first propeller of LRP5, a region of importance for binding inhibitory proteins. Thereby, ADO1 cannot be regarded as a classical form of osteopetrosis but must now be considered a disease of LRP5 activation. ADO (Albers-Schönberg disease, or previously ADO2) is characterized by increased number of osteoclasts and a defect in the chloride transport system (ClC-7) of importance for acidification of the resorption lacuna (a form of Chloride Channel 7 Deficiency Osteopetrosis). Ex vivo studies of osteoclasts from ADO have shown that cells do form normally but have reduced resorption capacity and an expanded life span. Bone formation seems normal despite decreased osteoclast function. Uncoupling of formation from resorption makes ADO of interest for new strategies for treatment of osteoporosis. Recent studies have integrated bone metabolism in whole-body energy homeostasis. Patients with ADO may have decreased insulin levels indicating importance beyond bone metabolism. There seems to be a paradigm shift in the treatment of osteoporosis. Targeting ClC-7 might introduce a new principle of dual action. Drugs affecting ClC-7 could be antiresorptive, still allowing ongoing bone formation. Inversely, drugs affecting the inhibitory site of LRP5 might stimulate bone formation and inhibit resorption. Thereby, these studies have highlighted several intriguing treatment possibilities, employing novel modes of action, which could provide benefits to the treatment of osteoporosis.
Collapse
Affiliation(s)
- Jens Bollerslev
- Section of Specialized Endocrinology, Medical Clinic B, Rikshospitalet, Oslo University Hospital, N-0027 Oslo, Norway.
| | | | | | | | | |
Collapse
|
39
|
Yokota H, Hamamura K, Chen A, Dodge TR, Tanjung N, Abedinpoor A, Zhang P. Effects of salubrinal on development of osteoclasts and osteoblasts from bone marrow-derived cells. BMC Musculoskelet Disord 2013; 14:197. [PMID: 23816340 PMCID: PMC3711788 DOI: 10.1186/1471-2474-14-197] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 06/14/2013] [Indexed: 11/26/2022] Open
Abstract
Background Osteoporosis is a skeletal disease leading to an increased risk of bone fracture. Using a mouse osteoporosis model induced by administration of a receptor activator of nuclear factor kappa-B ligand (RANKL), salubrinal was recently reported as a potential therapeutic agent. To evaluate the role of salubrinal in cellular fates as well as migratory and adhesive functions of osteoclast/osteoblast precursors, we examined the development of primary bone marrow-derived cells in the presence and absence of salubrinal. We addressed a question: are salubrinal’s actions more potent to the cells isolated from the osteoporotic mice than those isolated from the control mice? Methods Using the RANKL-injected and control mice, bone marrow-derived cells were harvested. Osteoclastogenesis was induced by macrophage-colony stimulating factor and RANKL, while osteoblastogenesis was driven by dexamethasone, ascorbic acid, and β-glycerophosphate. Results The results revealed that salubrinal suppressed the numbers of colony forming-unit (CFU)-granulocyte/macrophages and CFU-macrophages, as well as formation of mature osteoclasts in a dosage-dependent manner. Salubrinal also suppressed migration and adhesion of pre-osteoclasts and increased the number of CFU-osteoblasts. Salubrinal was more effective in exerting its effects in the cells isolated from the RANKL-injected mice than the control. Consistent with cellular fates and functions, salubrinal reduced the expression of nuclear factor of activated T cells c1 (NFATc1) as well as tartrate-resistant acid phosphatase. Conclusions The results support the notion that salubrinal exhibits significant inhibition of osteoclastogenesis as well as stimulation of osteoblastogenesis in bone marrow-derived cells, and its efficacy is enhanced in the cells harvested from the osteoporotic bone samples.
Collapse
Affiliation(s)
- Hiroki Yokota
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, 723 West Michigan Street, SL220, Indianapolis, IN 46202, USA.
| | | | | | | | | | | | | |
Collapse
|
40
|
RANKL cytokine: from pioneer of the osteoimmunology era to cure for a rare disease. Clin Dev Immunol 2013; 2013:412768. [PMID: 23762088 PMCID: PMC3671266 DOI: 10.1155/2013/412768] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Accepted: 04/23/2013] [Indexed: 12/13/2022]
Abstract
Since its identification, the RANKL cytokine has been demonstrated to play a crucial role in bone homeostasis and lymphoid tissue organization. Genetic defects impairing its function lead to a peculiar form of autosomal recessive osteopetrosis (ARO), a rare genetic bone disease presenting early in life and characterized by increased bone density due to failure in bone resorption by the osteoclasts. Hematopoietic stem cell transplantation (HSCT) is the only option for the majority of patients affected by this life-threatening disease. However, the RANKL-dependent ARO does not gain any benefit from this approach, because the genetic defect is not intrinsic to the hematopoietic osteoclast lineage but rather to the mesenchymal one. Of note, we recently provided proof of concept of the efficacy of a pharmacological RANKL-based therapy to cure this form of the disease. Here we provide an overview of the diverse roles of RANKL in the bone and immune systems and review the clinical features of RANKL-deficient ARO patients and the results of our preclinical studies. We emphasize that these patients present a continuous worsening of the disease in the absence of a cure and strongly wish that the therapy we propose will be further developed.
Collapse
|