Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Nat Immunol ; 25(1): 102-116, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38012418

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapies have successfully treated hematological malignancies. Macrophages have also gained attention as an immunotherapy owing to their immunomodulatory capacity and ability to infiltrate solid tumors and phagocytize tumor cells. The first-generation CD3ζ-based CAR-macrophages could phagocytose tumor cells in an antigen-dependent manner. Here we engineered induced pluripotent stem cell-derived macrophages (iMACs) with toll-like receptor 4 intracellular toll/IL-1R (TIR) domain-containing CARs resulting in a markedly enhanced antitumor effect over first-generation CAR-macrophages. Moreover, the design of a tandem CD3ζ-TIR dual signaling CAR endows iMACs with both target engulfment capacity and antigen-dependent M1 polarization and M2 resistance in a nuclear factor kappa B (NF-κB)-dependent manner, as well as the capacity to modulate the tumor microenvironment. We also outline a mechanism of tumor cell elimination by CAR-induced efferocytosis against tumor cell apoptotic bodies. Taken together, we provide a second-generation CAR-iMAC with an ability for orthogonal phagocytosis and polarization and superior antitumor functions in treating solid tumors relative to first-generation CAR-macrophages.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Humans , Receptors, Antigen, T-Cell , T-Lymphocytes , Cell Line, Tumor , Receptors, Chimeric Antigen/genetics , Immunotherapy, Adoptive/methods , Macrophages/pathology , Tumor Microenvironment
3.
Genome Res ; 31(5): 900-909, 2021 05.
Article in English | MEDLINE | ID: mdl-33795333

ABSTRACT

Targeted mutagenesis in model organisms is key for gene functional annotation and biomedical research. Despite technological advances in gene editing by the CRISPR-Cas9 systems, rapid and efficient introduction of site-directed mutations remains a challenge in large animal models. Here, we developed a robust and flexible insertional mutagenesis strategy, homology-independent targeted trapping (HIT-trapping), which is generic and can efficiently target-trap an endogenous gene of interest independent of homology arm and embryonic stem cells. Further optimization and equipping the HIT-trap donor with a site-specific DNA inversion mechanism enabled one-step generation of reversible and conditional alleles in a single experiment. As a proof of concept, we successfully created mutant alleles for 21 disease-related genes in primary porcine fibroblasts with an average knock-in frequency of 53.2%, a great improvement over previous approaches. The versatile HIT-trapping strategy presented here is expected to simplify the targeted generation of mutant alleles and facilitate large-scale mutagenesis in large mammals such as pigs.


Subject(s)
Alleles , CRISPR-Cas Systems , Gene Editing , Animals , Mutagenesis, Insertional , Mutation , Swine
4.
Proc Natl Acad Sci U S A ; 114(4): 722-727, 2017 01 24.
Article in English | MEDLINE | ID: mdl-28062688

ABSTRACT

CRISPR/Cas9 is becoming an increasingly important tool to functionally annotate genomes. However, because genome-wide CRISPR libraries are mostly constructed in lentiviral vectors, in vivo applications are severely limited as a result of difficulties in delivery. Here, we examined the piggyBac (PB) transposon as an alternative vehicle to deliver a guide RNA (gRNA) library for in vivo screening. Although tumor induction has previously been achieved in mice by targeting cancer genes with the CRISPR/Cas9 system, in vivo genome-scale screening has not been reported. With our PB-CRISPR libraries, we conducted an in vivo genome-wide screen in mice and identified genes mediating liver tumorigenesis, including known and unknown tumor suppressor genes (TSGs). Our results demonstrate that PB can be a simple and nonviral choice for efficient in vivo delivery of CRISPR libraries.


Subject(s)
CRISPR-Cas Systems/genetics , Carcinogenesis/genetics , Animals , DNA Transposable Elements/genetics , Gene Library , Genes, Tumor Suppressor/physiology , Genetic Engineering/methods , Genome/genetics , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , RNA, Guide, Kinetoplastida/genetics
5.
Nat Commun ; 15(1): 1256, 2024 Feb 10.
Article in English | MEDLINE | ID: mdl-38341436

ABSTRACT

LIN28A is important in somatic reprogramming and pluripotency regulation. Although previous studies addressed that LIN28A can repress let-7 microRNA maturation in the cytoplasm, few focused on its role within the nucleus. Here, we show that the nucleolus-localized LIN28A protein undergoes liquid-liquid phase separation (LLPS) in mouse embryonic stem cells (mESCs) and in vitro. The RNA binding domains (RBD) and intrinsically disordered regions (IDR) of LIN28A contribute to LIN28A and the other nucleolar proteins' phase-separated condensate establishment. S120A, S200A and R192G mutations in the IDR result in subcellular mislocalization of LIN28A and abnormal nucleolar phase separation. Moreover, we find that the naive-to-primed pluripotency state conversion and the reprogramming are associated with dynamic nucleolar remodeling, which depends on LIN28A's phase separation capacity, because the LIN28A IDR point mutations abolish its role in regulating nucleolus and in these cell fate decision processes, and an exogenous IDR rescues it. These findings shed light on the nucleolar function in pluripotent stem cell states and on a non-canonical RNA-independent role of LIN28A in phase separation and cell fate decisions.


Subject(s)
Phase Separation , Pluripotent Stem Cells , RNA-Binding Proteins , Animals , Mice , Cell Differentiation/genetics , Cell Nucleolus/metabolism , Pluripotent Stem Cells/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
6.
Cells ; 11(10)2022 05 16.
Article in English | MEDLINE | ID: mdl-35626689

ABSTRACT

The Chimeric antigen receptor (CAR)-T cell therapy has made inroads in treating hematological malignancies. Nonetheless, there are still multiple hurdles in CAR-T cell therapy for solid tumors. Primary CAR-expressing macrophage cells (CAR-Ms) and induced pluripotent stem cells (iPSCs)-derived CAR-expressing macrophage cells (CAR-iMacs) have emerged as attractive alternatives in our quest for an efficient and inexpensive approach for tumor immune cell therapy. In this review, we list the current state of development of human CAR-macrophages and provide an overview of the crucial functions of human CAR-macrophages in the field of tumor immune cell therapy.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Humans , Macrophages/metabolism , Neoplasms/pathology , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes
SELECTION OF CITATIONS
SEARCH DETAIL