Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
Hum Gene Ther ; 2024 May 03.
Article in English | MEDLINE | ID: mdl-38661537

ABSTRACT

The development of bone-targeting drug delivery systems holds immense promise for improving the treatment of skeletal diseases. By precisely delivering therapeutic agents to the affected areas of bone, these strategies can enhance drug efficacy, minimize off-target effects, and promote patient adherence, ultimately leading to improved treatment outcomes and an enhanced quality of life for patients. This review aims to provide an overview of the current state of affinity-based bone-targeting agents and recent breakthroughs in innovative bone-targeting adeno-associated virus (AAV) strategies to treat skeletal diseases in mice. In particular, this review will delve into advanced AAV engineering, including AAV serotype selection for bone targeting and capsid modifications for bone-specific tropism. Additionally, we will highlight recent advancements in AAV-mediated gene therapy for skeletal diseases and discuss challenges and future directions of this promising therapeutic approach.

2.
J Bone Miner Res ; 39(2): 106-115, 2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38477743

ABSTRACT

Artificial intelligence (AI) chatbots utilizing large language models (LLMs) have recently garnered significant interest due to their ability to generate humanlike responses to user inquiries in an interactive dialog format. While these models are being increasingly utilized to obtain medical information by patients, scientific and medical providers, and trainees to address biomedical questions, their performance may vary from field to field. The opportunities and risks these chatbots pose to the widespread understanding of skeletal health and science are unknown. Here we assess the performance of 3 high-profile LLM chatbots, Chat Generative Pre-Trained Transformer (ChatGPT) 4.0, BingAI, and Bard, to address 30 questions in 3 categories: basic and translational skeletal biology, clinical practitioner management of skeletal disorders, and patient queries to assess the accuracy and quality of the responses. Thirty questions in each of these categories were posed, and responses were independently graded for their degree of accuracy by four reviewers. While each of the chatbots was often able to provide relevant information about skeletal disorders, the quality and relevance of these responses varied widely, and ChatGPT 4.0 had the highest overall median score in each of the categories. Each of these chatbots displayed distinct limitations that included inconsistent, incomplete, or irrelevant responses, inappropriate utilization of lay sources in a professional context, a failure to take patient demographics or clinical context into account when providing recommendations, and an inability to consistently identify areas of uncertainty in the relevant literature. Careful consideration of both the opportunities and risks of current AI chatbots is needed to formulate guidelines for best practices for their use as source of information about skeletal health and biology.


Artificial intelligence chatbots are increasingly used as a source of information in health care and research settings due to their accessibility and ability to summarize complex topics using conversational language. However, it is still unclear whether they can provide accurate information for questions related to the medicine and biology of the skeleton. Here, we tested the performance of three prominent chatbots­ChatGPT, Bard, and BingAI­by tasking them with a series of prompts based on well-established skeletal biology concepts, realistic physician­patient scenarios, and potential patient questions. Despite their similarities in function, differences in the accuracy of responses were observed across the three different chatbot services. While in some contexts, chatbots performed well, and in other cases, strong limitations were observed, including inconsistent consideration of clinical context and patient demographics, occasionally providing incorrect or out-of-date information, and citation of inappropriate sources. With careful consideration of their current weaknesses, artificial intelligence chatbots offer the potential to transform education on skeletal health and science.


Subject(s)
Artificial Intelligence , Bone and Bones , Humans , Bone and Bones/physiology , Bone Diseases/therapy
3.
Hum Gene Ther ; 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38534217

ABSTRACT

Adeno-associated viral (AAV) vectors have emerged as crucial tools in advancing gene therapy for skeletal diseases, offering the potential for sustained expression with low postinfection immunogenicity and pathogenicity. Preclinical studies support both the therapeutic efficacy and safety of these vectors, illustrating the promise of AAV-mediated gene therapy. Emerging technologies and innovations in AAV-mediated gene therapy strategies, such as gene addition, gene replacement, gene silencing, and gene editing, offer new approaches to clinical application. Recently, the increasing preclinical applications of AAV to rare skeletal diseases, such as fibrodysplasia ossificans progressiva (FOP) and osteogenesis imperfecta (OI), and prevalent bone diseases, such as osteoporosis, bone fracture, critical-sized bone defects, and osteoarthritis, have been reported. Despite existing limitations in clinical use, such as high cost and safety, the AAV-mediated gene transfer platform is a promising approach to deliver therapeutic gene(s) to the skeleton to treat skeletal disorders, including those otherwise intractable by other therapeutic approaches. This review provides a comprehensive overview of the therapeutic advancements, challenges, limitations, and solutions within AAV-based gene therapy for prevalent and rare skeletal diseases.

4.
Protein Cell ; 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38442300

ABSTRACT

Tissue-resident stem cells are essential for development and repair, and in the skeleton this function is fulfilled by recently identified skeletal stem cells (SSCs). However, recent work has identified that SSCs are not monolithic, with long bones, craniofacial sites, and the spine being formed by distinct stem cells. Recent studies have utilized techniques such as fluorescence-activated cell sorting (FACS), lineage tracing and single-cell sequencing to investigate the involvement of SSCs in bone development, homeostasis and disease. These investigations have allowed researchers to map the lineage commitment trajectory of SSCs in different parts of the body and at different time points. Furthermore, recent studies have shed light on the characteristics of SSCs in both physiological and pathological conditions. This review focuses on discussing the spatiotemporal distribution of SSCs and enhancing our understanding of the diversity and plasticity of SSCs by summarizing recent discoveries.

5.
Adv Sci (Weinh) ; 11(18): e2303752, 2024 May.
Article in English | MEDLINE | ID: mdl-38311573

ABSTRACT

Loss of refreshment in nucleus pulposus (NP) cellularity leads to intervertebral disc (IVD) degeneration. Nevertheless, the cellular sequence of NP cell differentiation remains unclear, although an increasing body of literature has identified markers of NP progenitor cells (NPPCs). Notably, due to their fragility, the physical enrichment of NP-derived cells has limited conventional transcriptomic approaches in multiple studies. To overcome this limitation, a spatially resolved transcriptional atlas of the mouse IVD is generated via the 10x Genomics Visium platform dividing NP spots into two clusters. Based on this, most reported NPPC-markers, including Cathepsin K (Ctsk), are rare and predominantly located within the NP-outer subset. Cell lineage tracing further evidence that a small number of Ctsk-expressing cells generate the entire adult NP tissue. In contrast, Tie2, which has long suggested labeling NPPCs, is actually neither expressed in NP subsets nor labels NPPCs and their descendants in mouse models; consistent with this, an in situ sequencing (ISS) analysis validated the absence of Tie2 in NP tissue. Similarly, no Tie2-cre-mediated labeling of NPPCs is observed in an IVD degenerative mouse model. Altogether, in this study, the first spatial transcriptomic map of the IVD is established, thereby providing a public resource for bone biology.


Subject(s)
Nucleus Pulposus , Stem Cells , Transcriptome , Animals , Mice , Nucleus Pulposus/metabolism , Nucleus Pulposus/cytology , Stem Cells/metabolism , Transcriptome/genetics , Cell Differentiation/genetics , Intervertebral Disc Degeneration/genetics , Intervertebral Disc Degeneration/metabolism , Gene Expression Profiling/methods , Disease Models, Animal
6.
J Orthop Res ; 42(6): 1276-1282, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38245845

ABSTRACT

Large language model (LLM) chatbots possess a remarkable capacity to synthesize complex information into concise, digestible summaries across a wide range of orthopedic subject matter. As LLM chatbots become widely available they will serve as a powerful, accessible resource that patients, clinicians, and researchers may reference to obtain information about orthopedic science and clinical management. Here, we examined the performance of three well-known and easily accessible chatbots-ChatGPT, Bard, and Bing AI-in responding to inquiries relating to clinical management and orthopedic concepts. Although all three chatbots were found to be capable of generating relevant responses, ChatGPT outperformed Bard and BingAI in each category due to its ability to provide accurate and complete responses to orthopedic queries. Despite their promising applications in clinical management, shortcomings observed included incomplete responses, lack of context, and outdated information. Nonetheless, the ability for these LLM chatbots to address these inquires has largely yet to be evaluated and will be critical for understanding the risks and opportunities of LLM chatbots in orthopedics.


Subject(s)
Orthopedics , Humans
7.
J Bone Joint Surg Am ; 106(8): 735-745, 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38194481

ABSTRACT

BACKGROUND: Multiple animal models have previously been utilized to investigate anterior fusion techniques, but a mouse model has yet to be developed. The purpose of this study was to develop murine anterior interbody and posterolateral fusion techniques. METHODS: Mice underwent either anterior interbody or posterolateral spinal fusion. A protocol was developed for both procedures, including a description of the relevant anatomy. Samples were subjected to micro-computed tomography to assess fusion success and underwent biomechanical testing with use of 4-point bending. Lastly, samples were fixed and embedded for histologic evaluation. RESULTS: Surgical techniques for anterior interbody and posterolateral fusion were developed. The fusion rate was 83.3% in the anterior interbody model and 100% in the posterolateral model. Compared with a control, the posterolateral model exhibited a greater elastic modulus. Histologic analysis demonstrated endochondral ossification between bridging segments, further confirming the fusion efficacy in both models. CONCLUSIONS: The murine anterior interbody and posterolateral fusion models are efficacious and provide an ideal platform for studying the molecular and cellular mechanisms mediating spinal fusion. CLINICAL RELEVANCE: Given the extensive genetic tools available in murine disease models, use of fusion models such as ours can enable determination of the underlying genetic pathways involved in spinal fusion.


Subject(s)
Lumbar Vertebrae , Spinal Fusion , Animals , Mice , Lumbar Vertebrae/surgery , Spinal Fusion/methods , X-Ray Microtomography , Osteogenesis , Disease Models, Animal
8.
Nat Commun ; 15(1): 363, 2024 Jan 08.
Article in English | MEDLINE | ID: mdl-38191471

ABSTRACT

In the complex tumor microenvironment (TME), mesenchymal cells are key players, yet their specific roles in prostate cancer (PCa) progression remain to be fully deciphered. This study employs single-cell RNA sequencing to delineate molecular changes in tumor stroma that influence PCa progression and metastasis. Analyzing mesenchymal cells from four genetically engineered mouse models (GEMMs) and correlating these findings with human tumors, we identify eight stromal cell populations with distinct transcriptional identities consistent across both species. Notably, stromal signatures in advanced mouse disease reflect those in human bone metastases, highlighting periostin's role in invasion and differentiation. From these insights, we derive a gene signature that predicts metastatic progression in localized disease beyond traditional Gleason scores. Our results illuminate the critical influence of stromal dynamics on PCa progression, suggesting new prognostic tools and therapeutic targets.


Subject(s)
Mesenchymal Stem Cells , Prostatic Neoplasms , Humans , Male , Animals , Mice , Prostatic Neoplasms/genetics , Prostate , Stromal Cells , Cell Differentiation , Tumor Microenvironment/genetics
9.
Int J Mol Sci ; 24(23)2023 Nov 29.
Article in English | MEDLINE | ID: mdl-38069267

ABSTRACT

Chronic inflammation contributes to the development of skeletal disorders in patients with systemic lupus erythematosus (SLE). Activation of the host immune response stimulates osteoclast activity, which in turn leads to bone loss. Regenerating bone in the inflammatory microenvironments of SLE patients with critical bone defects remains a great challenge. In this study, we utilized lipopolysaccharide (LPS) to imitate locally and systemically pathogenic bacterial infection and examined the bone regeneration performance of LPS-associated mandibular and tibial bone regeneration impairment in FcγRIIB-/- mice. Our results indicated that a loss of FcγRIIB alleviates bone regeneration in both mandibles and tibiae. After LPS induction, FcγRIIB-/- mice were susceptible to impaired fracture healing in tibial and mandibular bones. LPS decreased the mineralization to collagen ratio in FcγRIIB-/- mice, indicating a mineralization defect during bone repair. An osteoblast-associated gene (Col1a1) was attenuated in FcγRIIB-deficient mice, whereas Bglap, Hhip, and Creb5 were further downregulated with LPS treatment in FcγRIIB-/- mice compared to FcγRIIB-/- mice. Alpl and Bglap expression was dcreased in osteoblasts derived from bone chips. An osteoclast-associated gene, Tnfsf11/Tnfrsf11 ratio, ewas increased in LPS-induced FcγRIIB-/- mice and in vitro. Furthermore, systemic LPS was relatively potent in stimulating production of pro-inflammatory cytokines including TNF-α, IL-6, and MCP-1 in FcγRIIB-/- mice compared to FcγRIIB-/- mice. The levels of TNF-α, IFN-ß, IL-1α, and IL-17A were increased, whereas IL-10 and IL-23 were decreased in FcγRIIB-/- mice treated locally with LPS. These findings suggest that both local and systemic LPS burden can exacerbate bone regeneration impairment, delay mineralization and skeletal repair, and induce inflammation in SLE patients.


Subject(s)
Lipopolysaccharides , Lupus Erythematosus, Systemic , Animals , Mice , Inflammation , Lipopolysaccharides/toxicity , Osteoclasts , Tumor Necrosis Factor-alpha
10.
Proc Natl Acad Sci U S A ; 120(46): e2312677120, 2023 Nov 14.
Article in English | MEDLINE | ID: mdl-37931101

ABSTRACT

We have previously reported that the cortical bone thinning seen in mice lacking the Wnt signaling antagonist Sfrp4 is due in part to impaired periosteal apposition. The periosteum contains cells which function as a reservoir of stem cells and contribute to cortical bone expansion, homeostasis, and repair. However, the local or paracrine factors that govern stem cells within the periosteal niche remain elusive. Cathepsin K (Ctsk), together with additional stem cell surface markers, marks a subset of periosteal stem cells (PSCs) which possess self-renewal ability and inducible multipotency. Sfrp4 is expressed in periosteal Ctsk-lineage cells, and Sfrp4 global deletion decreases the pool of PSCs, impairs their clonal multipotency for differentiation into osteoblasts and chondrocytes and formation of bone organoids. Bulk RNA sequencing analysis of Ctsk-lineage PSCs demonstrated that Sfrp4 deletion down-regulates signaling pathways associated with skeletal development, positive regulation of bone mineralization, and wound healing. Supporting these findings, Sfrp4 deletion hampers the periosteal response to bone injury and impairs Ctsk-lineage periosteal cell recruitment. Ctsk-lineage PSCs express the PTH receptor and PTH treatment increases the % of PSCs, a response not seen in the absence of Sfrp4. Importantly, in the absence of Sfrp4, PTH-dependent increase in cortical thickness and periosteal bone formation is markedly impaired. Thus, this study provides insights into the regulation of a specific population of periosteal cells by a secreted local factor, and shows a central role for Sfrp4 in the regulation of Ctsk-lineage periosteal stem cell differentiation and function.


Subject(s)
Osteogenesis , Stem Cell Niche , Mice , Animals , Cathepsin K/metabolism , Periosteum/metabolism , Cell Differentiation/genetics , Wnt Signaling Pathway , Proto-Oncogene Proteins/metabolism
11.
Cells ; 12(20)2023 10 19.
Article in English | MEDLINE | ID: mdl-37887332

ABSTRACT

Bone morphogenetic protein (BMP) gene delivery to Lewis rat lumbar intervertebral discs (IVDs) drives bone formation anterior and external to the IVD, suggesting the IVD is inhospitable to osteogenesis. This study was designed to determine if IVD destruction with a proteoglycanase, and/or generating an IVD blood supply by gene delivery of an angiogenic growth factor, could render the IVD permissive to intra-discal BMP-driven osteogenesis and fusion. Surgical intra-discal delivery of naïve or gene-programmed cells (BMP2/BMP7 co-expressing or VEGF165 expressing) +/- purified chondroitinase-ABC (chABC) in all permutations was performed between lumbar 4/5 and L5/6 vertebrae, and radiographic, histology, and biomechanics endpoints were collected. Follow-up anti-sFlt Western blotting was performed. BMP and VEGF/BMP treatments had the highest stiffness, bone production and fusion. Bone was induced anterior to the IVD, and was not intra-discal from any treatment. chABC impaired BMP-driven osteogenesis, decreased histological staining for IVD proteoglycans, and made the IVD permissive to angiogenesis. A soluble fragment of VEGF Receptor-1 (sFlt) was liberated from the IVD matrix by incubation with chABC, suggesting dysregulation of the sFlt matrix attachment is a possible mechanism for the chABC-mediated IVD angiogenesis we observed. Based on these results, the IVD can be manipulated to foster vascular invasion, and by extension, possibly osteogenesis.


Subject(s)
Intervertebral Disc , Nucleus Pulposus , Rats , Animals , Nucleus Pulposus/metabolism , Vascular Endothelial Growth Factor A/metabolism , Rats, Inbred Lew , Intervertebral Disc/pathology , Proteoglycans/metabolism
12.
Clin Chem ; 69(11): 1238-1246, 2023 11 02.
Article in English | MEDLINE | ID: mdl-37664912

ABSTRACT

BACKGROUND: Artificial intelligence (AI) conversational agents, or chatbots, are computer programs designed to simulate human conversations using natural language processing. They offer diverse functions and applications across an expanding range of healthcare domains. However, their roles in laboratory medicine remain unclear, as their accuracy, repeatability, and ability to interpret complex laboratory data have yet to be rigorously evaluated. CONTENT: This review provides an overview of the history of chatbots, two major chatbot development approaches, and their respective advantages and limitations. We discuss the capabilities and potential applications of chatbots in healthcare, focusing on the laboratory medicine field. Recent evaluations of chatbot performance are presented, with a special emphasis on large language models such as the Chat Generative Pre-trained Transformer in response to laboratory medicine questions across different categories, such as medical knowledge, laboratory operations, regulations, and interpretation of laboratory results as related to clinical context. We analyze the causes of chatbots' limitations and suggest research directions for developing more accurate, reliable, and manageable chatbots for applications in laboratory medicine. SUMMARY: Chatbots, which are rapidly evolving AI applications, hold tremendous potential to improve medical education, provide timely responses to clinical inquiries concerning laboratory tests, assist in interpreting laboratory results, and facilitate communication among patients, physicians, and laboratorians. Nevertheless, users should be vigilant of existing chatbots' limitations, such as misinformation, inconsistencies, and lack of human-like reasoning abilities. To be effectively used in laboratory medicine, chatbots must undergo extensive training on rigorously validated medical knowledge and be thoroughly evaluated against standard clinical practice.


Subject(s)
Clinical Laboratory Services , Medicine , Humans , Laboratories, Clinical , Artificial Intelligence , Laboratories
13.
Nature ; 621(7980): 804-812, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37730988

ABSTRACT

Craniosynostosis is a group of disorders of premature calvarial suture fusion. The identity of the calvarial stem cells (CSCs) that produce fusion-driving osteoblasts in craniosynostosis remains poorly understood. Here we show that both physiologic calvarial mineralization and pathologic calvarial fusion in craniosynostosis reflect the interaction of two separate stem cell lineages; a previously identified cathepsin K (CTSK) lineage CSC1 (CTSK+ CSC) and a separate discoidin domain-containing receptor 2 (DDR2) lineage stem cell (DDR2+ CSC) that we identified in this study. Deletion of Twist1, a gene associated with craniosynostosis in humans2,3, solely in CTSK+ CSCs is sufficient to drive craniosynostosis in mice, but the sites that are destined to fuse exhibit an unexpected depletion of CTSK+ CSCs and a corresponding expansion of DDR2+ CSCs, with DDR2+ CSC expansion being a direct maladaptive response to CTSK+ CSC depletion. DDR2+ CSCs display full stemness features, and our results establish the presence of two distinct stem cell lineages in the sutures, with both populations contributing to physiologic calvarial mineralization. DDR2+ CSCs mediate a distinct form of endochondral ossification without the typical haematopoietic marrow formation. Implantation of DDR2+ CSCs into suture sites is sufficient to induce fusion, and this phenotype was prevented by co-transplantation of CTSK+ CSCs. Finally, the human counterparts of DDR2+ CSCs and CTSK+ CSCs display conserved functional properties in xenograft assays. The interaction between these two stem cell populations provides a new biologic interface for the modulation of calvarial mineralization and suture patency.


Subject(s)
Craniosynostoses , Humans , Mice , Animals , Craniosynostoses/genetics , Osteogenesis , Cell Lineage , Phenotype , Stem Cells
14.
Nature ; 621(7979): 602-609, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37704733

ABSTRACT

Vertebral bone is subject to a distinct set of disease processes from long bones, including a much higher rate of solid tumour metastases1-4. The basis for this distinct biology of vertebral bone has so far remained unknown. Here we identify a vertebral skeletal stem cell (vSSC) that co-expresses ZIC1 and PAX1 together with additional cell surface markers. vSSCs display formal evidence of stemness, including self-renewal, label retention and sitting at the apex of their differentiation hierarchy. vSSCs are physiologic mediators of vertebral bone formation, as genetic blockade of the ability of vSSCs to generate osteoblasts results in defects in the vertebral neural arch and body. Human counterparts of vSSCs can be identified in vertebral endplate specimens and display a conserved differentiation hierarchy and stemness features. Multiple lines of evidence indicate that vSSCs contribute to the high rates of vertebral metastatic tropism observed in breast cancer, owing in part to increased secretion of the novel metastatic trophic factor MFGE8. Together, our results indicate that vSSCs are distinct from other skeletal stem cells and mediate the unique physiology and pathology of vertebrae, including contributing to the high rate of vertebral metastasis.


Subject(s)
Breast Neoplasms , Cell Lineage , Neoplasm Metastasis , Spine , Stem Cells , Humans , Breast Neoplasms/pathology , Cell Differentiation , Cell Self Renewal , Neoplasm Metastasis/pathology , Osteoblasts/cytology , Osteoblasts/pathology , Spine/cytology , Spine/pathology , Stem Cells/cytology , Stem Cells/metabolism , Stem Cells/pathology , Biomarkers
15.
Int J Mol Sci ; 24(13)2023 Jun 28.
Article in English | MEDLINE | ID: mdl-37445982

ABSTRACT

Transforming growth factor beta (TGF-ß) is a key factor mediating the intercellular crosstalk between the hematopoietic stem cells and their microenvironment. Here, we investigated the skeletal phenotype of transgenic mice expressing constitutively active TGF-ß receptor type I under the control of Mx1-Cre (Mx1;TßRICA mice). µCT analysis showed decreased cortical thickness, and cancellous bone volume in both femurs and mandibles. Histomorphometric analysis confirmed a decrease in cancellous bone volume due to increased osteoclast number and decreased osteoblast number. Primary osteoblasts showed decreased ALP and mineralization. Constitutive TßRI activation increased osteoclast differentiation. qPCR analysis showed that Tnfsf11/Tnfrsf11b ratio, Ctsk, Sufu, and Csf1 were increased whereas Runx2, Ptch1, and Ptch2 were decreased in Mx1;TßRICA femurs. Interestingly, Gli1, Wnt3a, Sp7, Alpl, Ptch1, Ptch2, and Shh mRNA expression were reduced whereas Tnfsf11/Tnfrsf11b ratio was increased in Mx1;TßRICA mandibles. Similarly, osteoclast-related genes were increased in Mx1;TßRICA osteoclasts whereas osteoblast-related genes were reduced in Mx1;TßRICA osteoblasts. Western blot analysis indicated that SMAD2 and SMAD3 phosphorylation was increased in Mx1;TßRICA osteoblasts, and SMAD3 phosphorylation was increased in Mx1;TßRICA osteoclasts. CTSK was increased while RUNX2 and PTCH1 was decreased in Mx1;TßRICA mice. Microindentation analysis indicated decreased hardness in Mx1;TßRICA mice. Our study indicated that Mx1;TßRICA mice were osteopenic by increasing osteoclast number and decreasing osteoblast number, possibly by suppressing Hedgehog signaling pathways.


Subject(s)
Bone Diseases, Metabolic , Core Binding Factor Alpha 1 Subunit , Mice , Animals , Mice, Transgenic , Core Binding Factor Alpha 1 Subunit/metabolism , Cell Differentiation , Hedgehog Proteins/metabolism , Osteoblasts/metabolism , Osteoclasts/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Bone Diseases, Metabolic/metabolism
17.
JBMR Plus ; 7(4): e10723, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37065630

ABSTRACT

Aldehyde dehydrogenase 2 (ALDH2) deficiency affects 35% to 45% of East Asians and 8% of the world population. ALDH2 is the second enzyme in the ethanol metabolism pathway. The common genetic variant ALDH2*2 allele has a glutamic acid-to-lysine substitution at position 487 (E487K) that reduces the enzyme activity, resulting in an accumulation of acetaldehyde after ethanol consumption. The ALDH2*2 allele is associated with increased risk of osteoporosis and hip fracture. Our prior study showed that administration of an adeno-associated virus (AAV) serotype rh.10 gene transfer vector expressing the human ALDH2 cDNA (AAVrh.10hALDH2) before initiation of ethanol consumption prevented bone loss in ALDH2-deficient homozygous knockin mice carrying the E487K mutation (Aldh2 E487K+/+). We hypothesized that AAVrh.10hALDH2 administration after establishment of osteopenia would be able to reverse bone loss due to ALDH2 deficiency and chronic ethanol consumption. To test this hypothesis, male and female Aldh2 E487K+/+ mice (n = 6) were given ethanol in the drinking water for 6 weeks to establish osteopenia and then administered AAVrh.10hALDH2 (1011 genome copies). Mice were evaluated for an additional 12 weeks. AAVrh.10hALDH2 administration after osteopenia was established corrected weight loss and locomotion phenotypes and, importantly, increased midshaft femur cortical bone thickness, the most important component of bone in the resistance to fractures, and showed a trend toward increased trabecular bone volume. AAVrh.10hALDH2 is a promising therapeutic for osteoporosis in ALDH2-deficient individuals. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.

18.
bioRxiv ; 2023 Apr 01.
Article in English | MEDLINE | ID: mdl-37034687

ABSTRACT

Alterations in tumor stroma influence prostate cancer progression and metastatic potential. However, the molecular underpinnings of this stromal-epithelial crosstalk are largely unknown. Here, we compare mesenchymal cells from four genetically engineered mouse models (GEMMs) of prostate cancer representing different stages of the disease to their wild-type (WT) counterparts by single-cell RNA sequencing (scRNA-seq) and, ultimately, to human tumors with comparable genotypes. We identified 8 transcriptionally and functionally distinct stromal populations responsible for common and GEMM-specific transcriptional programs. We show that stromal responses are conserved in mouse models and human prostate cancers with the same genomic alterations. We noted striking similarities between the transcriptional profiles of the stroma of murine models of advanced disease and those of of human prostate cancer bone metastases. These profiles were then used to build a robust gene signature that can predict metastatic progression in prostate cancer patients with localized disease and is also associated with progression-free survival independent of Gleason score. Taken together, this offers new evidence that stromal microenvironment mediates prostate cancer progression, further identifying tissue-based biomarkers and potential therapeutic targets of aggressive and metastatic disease.

19.
Bone ; 172: 116761, 2023 07.
Article in English | MEDLINE | ID: mdl-37030497

ABSTRACT

Often, disorders of impaired bone formation involve not only a cell intrinsic defect in the ability of osteoblasts to form bone, but moreover a broader dysfunction of the skeletal microenvironment that limits osteoblast activity. Developing approaches to osteoanabolic therapy that not only augment osteoblast activity but moreover correct this microenvironmental dysfunction may enable both more effective osteoanabolic therapies and also addressing a broader set of indications where vasculopathy or other forms microenvironment dysfunction feature prominently. We here review evidence that SHN3 acts as a suppressor of not only the cell intrinsic bone formation activity of osteoblasts, but moreover of the creation of a local osteoanabolic microenvironment. Mice lacking Schnurri3 (SHN3, HIVEP3) display a very robust increase in bone formation, that is due to de-repression of ERK pathway signaling in osteoblasts. In addition to loss of SHN3 augmenting the differentiation and bone formation activity of osteoblasts, loss of SHN3 increases secretion of SLIT3 by osteoblasts, which in a skeletal context acts as an angiogenic factor. Through this angiogenic activity, SLIT3 creates an osteoanabolic microenvironment, and accordingly treatment with SLIT3 can increase bone formation and enhance fracture healing. These features both validate vascular endothelial cells as a therapeutic target for disorders of low bone mass alongside the traditionally targeted osteoblasts and osteoclasts and indicate that targeting the SHN3/SLIT3 pathway provides a new mechanism to induce therapeutic osteoanabolic responses.


Subject(s)
DNA-Binding Proteins , Endothelial Cells , Mice , Animals , Endothelial Cells/metabolism , DNA-Binding Proteins/metabolism , Osteoclasts/metabolism , Bone and Bones/metabolism , Osteoblasts/metabolism , Osteogenesis/physiology , Cell Differentiation , Membrane Proteins/metabolism
20.
Nat Mater ; 22(3): 391-399, 2023 03.
Article in English | MEDLINE | ID: mdl-36864161

ABSTRACT

Medulloblastoma is the most common malignant paediatric brain tumour, with ~30% mediated by Sonic hedgehog signalling. Vismodegib-mediated inhibition of the Sonic hedgehog effector Smoothened inhibits tumour growth but causes growth plate fusion at effective doses. Here, we report a nanotherapeutic approach targeting endothelial tumour vasculature to enhance blood-brain barrier crossing. We use fucoidan-based nanocarriers targeting endothelial P-selectin to induce caveolin-1-dependent transcytosis and thus nanocarrier transport into the brain tumour microenvironment in a selective and active manner, the efficiency of which is increased by radiation treatment. In a Sonic hedgehog medulloblastoma animal model, fucoidan-based nanoparticles encapsulating vismodegib exhibit a striking efficacy and marked reduced bone toxicity and drug exposure to healthy brain tissue. Overall, these findings demonstrate a potent strategy for targeted intracranial pharmacodelivery that overcomes the restrictive blood-brain barrier to achieve enhanced tumour-selective penetration and has therapeutic implications for diseases within the central nervous system.


Subject(s)
Cerebellar Neoplasms , Medulloblastoma , Animals , Hedgehog Proteins , Blood-Brain Barrier , Caveolin 1 , P-Selectin , Transcytosis , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...