Page 143 - OR-1-2
P. 143

locus  notch   homolog   protein  1 ],  suggesting  experiments in drug development, with a strategic shift
                                              +
            endothelial-to-hematopoietic transition), and mesenchymal   toward “new approach methodologies,” such as artificial
            populations (pericytes [angiopoietin 1 ], vascular smooth   intelligence-based predictive models and organoid toxicity
                                           +
            muscle cells [actin alpha 2 ], and osteochondral precursors   testing. Organoids, as a core technology in this transition,
                                 +
            [paired related homeobox 1 ]). This high-resolution   exhibit distinct advantages in precision, efficiency, and
                                      +
            molecular map provides a foundation for analyzing cellular   ethical compliance, with demonstrated applications across
            interactions within the niche (Figure 1B).        multiple research and  development  scenarios.  The  high-
                                                              throughput potential of BMOs (e.g., 96-well plate culture)
            2. Paradigm shift: From static models to          supports diverse toxicity assessment studies, including
            dynamic simulation                                hematotoxicity screening, evaluation of synergistic

            Unlike  previous  organoids  modeling  yolk  sac  damaging effects of chemotherapeutic agents on HSCs
            hematopoiesis,   the  more  recently  developed  BMOs   and stromal cells, and validation of targeted therapies (e.g.,
                        8
            replicated key features of the aorta-gonad-mesonephros   testing the therapeutic efficacy of Janus kinase inhibitors in
            region via arterial endothelial differentiation. scRNA-seq   myelofibrosis-modeled organoids).
            showed HSCs expressing fetal liver-stage markers (e.g.,   By integrating gene editing with organoid technology,
            selectin L, Musashi RNA binding protein 2), indicating their   BMOs enable HSC expansion optimization through
            transitional state toward mature HSCs. This discovery offers   supplementation with retinoic acid or Hedgehog pathway
            insights into human HSC development and differentiation   agonists, thereby enhancing transplantation efficiency.
            trajectories.                                     Furthermore, constructing mutation-specific organoids
               Upon lipopolysaccharide stimulation, BMOs secreted   based on individual genetic profiles allows screening of
            interleukin (IL)-6/IL-8 within 4  h, with pre-neural   personalized therapeutic regimens, making patient-specific
            populations expanding 2.5-fold by 24 h. This rapid response   treatments feasible. 12
            mirrors in vivo “emergency hematopoiesis” and demonstrates
            niche-autonomous regulation of granulocyte differentiation   4. Conclusion
            without exogenous granulocyte colony-stimulating factor.   The  in  vitro reconstruction of the bone marrow niche
            This model enables dynamic studies of post-infection or   represents not only a technical achievement but also a
            chemotherapy-induced bone marrow regeneration.    paradigm shift from “reductionism” to “systems biology.”

               Clustered regularly interspaced short palindromic   Integrating spatial transcriptomics and live-cell imaging,
            repeats-engineered BMOs with VPS45 mutations exhibited   BMOs may decode spatiotemporal cellular crosstalk
            increased reticulin fiber deposition, expansion of alpha-  within niches, bridging molecular mechanisms to clinical
            smooth muscle actin  myofibroblasts, and collagen   translation. Future BMOs could serve as “ex vivo testbeds”
                                +
            accumulation, leading to hematopoietic defects and   for personalized medicine, offering novel strategies for the
            elevated apoptosis of mature neutrophils, consistent with   design of drug screening models, in vivo regeneration, and
            bone marrow biopsy phenotypes in  VPS45-deficient   precision therapies.
            patients (Figure  1C). Such “genotype-phenotype linkage   Acknowledgments
            models” may offer biopsy-free solutions for studying rare
            hematologic disorders. 9-11                       None.

            3. Expansion of applications: From basic          Funding
            research to clinical translation                  This work was supported by Natural Science Foundation of

            Leukemia cells rely on the bone marrow microenvironment   Shandong Province (grant no.: ZR2023MC125) and Marine
            to acquire drug resistance. By integrating patient-derived   Aquaculture Innovation and Entrepreneurship Consortium
            induced pluripotent stem cells, BMOs can be engineered   Project of Shandong Province (grant no.: YZ2024007).
            into “leukemia niche organoids” to investigate stroma-
            mediated drug resistance mechanisms (e.g., the role of   Conflict of interest
            the C-X-C motif chemokine ligand 12-C-X-C chemokine   The authors declare they have no competing interests.
            receptor type  4 axis in minimal residual disease) and
            evaluate immune cell interactions (e.g., infiltration and  Author contributions
            cytotoxic efficacy of chimeric antigen receptor T-cell   Conceptualization: Liangyu Guo, Panpan Pan
            therapy cells within organoids).                  Formal analysis: Yifan Xia
               On April 10, 2025, the  United States  Food and Drug   Investigation: All authors
            Administration announced a phased elimination of animal   Methodology: Liangyu Guo, Yifan Xia


            Volume 1 Issue 2 (2025)                         3                            doi: 10.36922/OR025110011
   138   139   140   141   142   143   144   145   146