Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 214
Filter
Add more filters

Publication year range
1.
Cell ; 177(2): 315-325.e14, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30929905

ABSTRACT

Transmission of malaria parasites occurs when a female Anopheles mosquito feeds on an infected host to acquire nutrients for egg development. How parasites are affected by oogenetic processes, principally orchestrated by the steroid hormone 20-hydroxyecdysone (20E), remains largely unknown. Here we show that Plasmodium falciparum development is intimately but not competitively linked to processes shaping Anopheles gambiae reproduction. We unveil a 20E-mediated positive correlation between egg and oocyst numbers; impairing oogenesis by multiple 20E manipulations decreases parasite intensities. These manipulations, however, accelerate Plasmodium growth rates, allowing sporozoites to become infectious sooner. Parasites exploit mosquito lipids for faster growth, but they do so without further affecting egg development. These results suggest that P. falciparum has adopted a non-competitive evolutionary strategy of resource exploitation to optimize transmission while minimizing fitness costs to its mosquito vector. Our findings have profound implications for currently proposed control strategies aimed at suppressing mosquito populations.


Subject(s)
Ecdysterone/metabolism , Host-Parasite Interactions/physiology , Malaria, Falciparum/parasitology , Animals , Anopheles/parasitology , Culicidae , Ecdysterone/physiology , Female , HEK293 Cells , Humans , Insect Vectors , Malaria/parasitology , Mice , Mosquito Vectors , NIH 3T3 Cells , Oogenesis/physiology , Plasmodium/metabolism , Plasmodium falciparum , Sporozoites , Steroids/metabolism
2.
Cell ; 150(4): 842-54, 2012 Aug 17.
Article in English | MEDLINE | ID: mdl-22901813

ABSTRACT

Due to genome instability, most cancers exhibit loss of regions containing tumor suppressor genes and collateral loss of other genes. To identify cancer-specific vulnerabilities that are the result of copy number losses, we performed integrated analyses of genome-wide copy number and RNAi profiles and identified 56 genes for which gene suppression specifically inhibited the proliferation of cells harboring partial copy number loss of that gene. These CYCLOPS (copy number alterations yielding cancer liabilities owing to partial loss) genes are enriched for spliceosome, proteasome, and ribosome components. One CYCLOPS gene, PSMC2, encodes an essential member of the 19S proteasome. Normal cells express excess PSMC2, which resides in a complex with PSMC1, PSMD2, and PSMD5 and acts as a reservoir protecting cells from PSMC2 suppression. Cells harboring partial PSMC2 copy number loss lack this complex and die after PSMC2 suppression. These observations define a distinct class of cancer-specific liabilities resulting from genome instability.


Subject(s)
Genes, Essential , Genomic Instability , Neoplasms/genetics , ATPases Associated with Diverse Cellular Activities , Animals , Cell Line, Tumor , Chromosome Deletion , Gene Dosage , Genes, Tumor Suppressor , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms/metabolism , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Transplantation, Heterologous
3.
Nature ; 595(7866): 289-294, 2021 07.
Article in English | MEDLINE | ID: mdl-34194041

ABSTRACT

The global decline in malaria has stalled1, emphasizing the need for vaccines that induce durable sterilizing immunity. Here we optimized regimens for chemoprophylaxis vaccination (CVac), for which aseptic, purified, cryopreserved, infectious Plasmodium falciparum sporozoites (PfSPZ) were inoculated under prophylactic cover with pyrimethamine (PYR) (Sanaria PfSPZ-CVac(PYR)) or chloroquine (CQ) (PfSPZ-CVac(CQ))-which kill liver-stage and blood-stage parasites, respectively-and we assessed vaccine efficacy against homologous (that is, the same strain as the vaccine) and heterologous (a different strain) controlled human malaria infection (CHMI) three months after immunization ( https://clinicaltrials.gov/ , NCT02511054 and NCT03083847). We report that a fourfold increase in the dose of PfSPZ-CVac(PYR) from 5.12 × 104 to 2 × 105 PfSPZs transformed a minimal vaccine efficacy (low dose, two out of nine (22.2%) participants protected against homologous CHMI), to a high-level vaccine efficacy with seven out of eight (87.5%) individuals protected against homologous and seven out of nine (77.8%) protected against heterologous CHMI. Increased protection was associated with Vδ2 γδ T cell and antibody responses. At the higher dose, PfSPZ-CVac(CQ) protected six out of six (100%) participants against heterologous CHMI three months after immunization. All homologous (four out of four) and heterologous (eight out of eight) infectivity control participants showed parasitaemia. PfSPZ-CVac(CQ) and PfSPZ-CVac(PYR) induced a durable, sterile vaccine efficacy against a heterologous South American strain of P. falciparum, which has a genome and predicted CD8 T cell immunome that differs more strongly from the African vaccine strain than other analysed African P. falciparum strains.


Subject(s)
Antibodies, Neutralizing/immunology , Liver/immunology , Liver/parasitology , Malaria Vaccines/immunology , Plasmodium falciparum/drug effects , Plasmodium falciparum/immunology , Vaccines, Attenuated/immunology , Adult , Animals , Antibody Formation/immunology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Life Cycle Stages/immunology , Malaria/blood , Malaria/immunology , Malaria/parasitology , Malaria/prevention & control , Malaria Vaccines/administration & dosage , Malaria Vaccines/adverse effects , Malaria Vaccines/chemistry , Male , Middle Aged , Plasmodium falciparum/growth & development , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Time Factors , Vaccination/adverse effects , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/chemistry
4.
Proc Natl Acad Sci U S A ; 121(11): e2307802121, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38437557

ABSTRACT

RNA interference (RNAi) therapeutics are an emerging class of medicines that selectively target mRNA transcripts to silence protein production and combat disease. Despite the recent progress, a generalizable approach for monitoring the efficacy of RNAi therapeutics without invasive biopsy remains a challenge. Here, we describe the development of a self-reporting, theranostic nanoparticle that delivers siRNA to silence a protein that drives cancer progression while also monitoring the functional activity of its downstream targets. Our therapeutic target is the transcription factor SMARCE1, which was previously identified as a key driver of invasion in early-stage breast cancer. Using a doxycycline-inducible shRNA knockdown in OVCAR8 ovarian cancer cells both in vitro and in vivo, we demonstrate that SMARCE1 is a master regulator of genes encoding proinvasive proteases in a model of human ovarian cancer. We additionally map the peptide cleavage profiles of SMARCE1-regulated proteases so as to design a readout for downstream enzymatic activity. To demonstrate the therapeutic and diagnostic potential of our approach, we engineered self-assembled layer-by-layer nanoparticles that can encapsulate nucleic acid cargo and be decorated with peptide substrates that release a urinary reporter upon exposure to SMARCE1-related proteases. In an orthotopic ovarian cancer xenograft model, theranostic nanoparticles were able to knockdown SMARCE1 which was in turn reported through a reduction in protease-activated urinary reporters. These LBL nanoparticles both silence gene products by delivering siRNA and noninvasively report on downstream target activity by delivering synthetic biomarkers to sites of disease, enabling dose-finding studies as well as longitudinal assessments of efficacy.


Subject(s)
Ovarian Neoplasms , Peptides , Humans , Female , RNA Interference , Peptides/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/therapy , Peptide Hydrolases , RNA, Small Interfering/genetics , Endopeptidases , Chromosomal Proteins, Non-Histone , DNA-Binding Proteins
5.
Annu Rev Cell Dev Biol ; 28: 385-410, 2012.
Article in English | MEDLINE | ID: mdl-23057744

ABSTRACT

Efforts in the interdisciplinary field of bioengineering have led to innovative methods for investigating the complexities of cell responses in vitro. These approaches have emphasized the reduction of complex multicomponent cellular microenvironments into distinct individual signals as a means to both (a) better construct mimics of in vivo microenvironments and (b) better deconstruct microenvironments to study them. Microtechnology tools, together with advances in biomaterials, have been fundamental to this progress by enabling the tightly controlled presentation of environmental cues and the improved systematic analysis of cellular perturbations. In this review, we describe bioengineering approaches for controlling and measuring cell-environmental interactions in vitro, including strategies for high-throughput analysis. We also describe the mechanistic insights gained by the use of these novel tools, with associated applications ranging from fundamental biological studies, in vitro modeling of in vivo processes, and cell-based therapies.


Subject(s)
Cell Culture Techniques , Cell Engineering/methods , Biomechanical Phenomena , Biomimetic Materials , Bioreactors , Cell Adhesion , Humans , Microfluidic Analytical Techniques , Tissue Array Analysis/methods
6.
Proc Natl Acad Sci U S A ; 119(25): e2121778119, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35696579

ABSTRACT

Community-acquired pneumonia (CAP) has been brought to the forefront of global health priorities due to the COVID-19 pandemic. However, classification of viral versus bacterial pneumonia etiology remains a significant clinical challenge. To this end, we have engineered a panel of activity-based nanosensors that detect the dysregulated activity of pulmonary host proteases implicated in the response to pneumonia-causing pathogens and produce a urinary readout of disease. The nanosensor targets were selected based on a human protease transcriptomic signature for pneumonia etiology generated from 33 unique publicly available study cohorts. Five mouse models of bacterial or viral CAP were developed to assess the ability of the nanosensors to produce etiology-specific urinary signatures. Machine learning algorithms were used to train diagnostic classifiers that could distinguish infected mice from healthy controls and differentiate those with bacterial versus viral pneumonia with high accuracy. This proof-of-concept diagnostic approach demonstrates a way to distinguish pneumonia etiology based solely on the host proteolytic response to infection.


Subject(s)
COVID-19 , Community-Acquired Infections , Gene Expression Profiling , Peptide Hydrolases , Pneumonia, Bacterial , Animals , Biosensing Techniques , COVID-19/genetics , Community-Acquired Infections/classification , Community-Acquired Infections/genetics , Community-Acquired Infections/virology , Disease Models, Animal , Humans , Machine Learning , Mice , Nanoparticles , Peptide Hydrolases/genetics , Pneumonia, Bacterial/classification , Pneumonia, Bacterial/genetics
7.
Proc Natl Acad Sci U S A ; 119(28): e2115867119, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35763565

ABSTRACT

Liver regeneration is a well-orchestrated process that is typically studied in animal models. Although previous animal studies have offered many insights into liver regeneration, human biology is less well understood. To this end, we developed a three-dimensional (3D) platform called structurally vascularized hepatic ensembles for analyzing regeneration (SHEAR) to model multiple aspects of human liver regeneration. SHEAR enables control over hemodynamic alterations to mimic those that occur during liver injury and regeneration and supports the administration of biochemical inputs such as cytokines and paracrine interactions with endothelial cells. We found that exposing the endothelium-lined channel to fluid flow led to increased secretion of regeneration-associated factors. Stimulation with relevant cytokines not only amplified the secretory response, but also induced cell-cycle entry of primary human hepatocytes (PHHs) embedded within the device. Further, we identified endothelial-derived mediators that are sufficient to initiate proliferation of PHHs in this context. Collectively, the data presented here underscore the importance of multicellular models that can recapitulate high-level tissue functions and demonstrate that the SHEAR device can be used to discover and validate conditions that promote human liver regeneration.


Subject(s)
Endothelial Cells , Hepatocytes , Liver Regeneration , Liver , Cell Culture Techniques, Three Dimensional , Cytokines , Humans , Liver/blood supply , Liver Regeneration/physiology
8.
Proc Natl Acad Sci U S A ; 117(24): 13428-13436, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32493746

ABSTRACT

The recent advent of immune checkpoint inhibitor (CPI) antibodies has revolutionized many aspects of cancer therapy, but the efficacy of these breakthrough therapeutics remains limited, as many patients fail to respond for reasons that still largely evade understanding. An array of studies in human patients and animal models has demonstrated that local signaling can generate strongly immunosuppressive microenvironments within tumors, and emerging evidence suggests that delivery of immunostimulatory molecules into tumors can have therapeutic effects. Nanoparticle formulations of these cargoes offer a promising way to maximize their delivery and to enhance the efficacy of checkpoint inhibitors. We developed a modular nanoparticle system capable of encapsulating an array of immunostimulatory oligonucleotides that, in some cases, greatly increase their potency to activate inflammatory signaling within immune cells in vitro. We hypothesized that these immunostimulatory nanoparticles could suppress tumor growth by activating similar signaling in vivo, and thereby also improve responsiveness to immune checkpoint inhibitor antibody therapies. We found that our engineered nanoparticles carrying a CpG DNA ligand of TLR9 can suppress tumor growth in several animal models of various cancers, resulting in an abscopal effect on distant tumors, and improving responsiveness to anti-CTLA4 treatment with combinatorial effects after intratumoral administration. Moreover, by incorporating tumor-homing peptides, immunostimulatory nucleotide-bearing nanoparticles facilitate antitumor efficacy after systemic intravenous (i.v.) administration.


Subject(s)
Adjuvants, Immunologic , Antineoplastic Agents, Immunological/therapeutic use , Immunotherapy/methods , Nanoparticles , Oligonucleotides/therapeutic use , Animals , CTLA-4 Antigen/antagonists & inhibitors , Cell Line , Drug Delivery Systems , Drug Therapy, Combination , Inflammation , Macrophages/drug effects , Macrophages/immunology , Mice , Nanoparticles/chemistry , Neoplasms, Experimental/therapy , Oligonucleotides/chemistry , Oligonucleotides/immunology
9.
Proc Natl Acad Sci U S A ; 117(3): 1678-1688, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31915293

ABSTRACT

Primary human hepatocytes (PHHs) are an essential tool for modeling drug metabolism and liver disease. However, variable plating efficiencies, short lifespan in culture, and resistance to genetic manipulation have limited their use. Here, we show that the pyrrolizidine alkaloid retrorsine improves PHH repopulation of chimeric mice on average 10-fold and rescues the ability of even poorly plateable donor hepatocytes to provide cells for subsequent ex vivo cultures. These mouse-passaged (mp) PHH cultures overcome the marked donor-to-donor variability of cryopreserved PHH and remain functional for months as demonstrated by metabolic assays and infection with hepatitis B virus and Plasmodium falciparum mpPHH can be efficiently genetically modified in culture, mobilized, and then recultured as spheroids or retransplanted to create highly humanized mice that carry a genetically altered hepatocyte graft. Together, these advances provide flexible tools for the study of human liver disease and evaluation of hepatocyte-targeted gene therapy approaches.


Subject(s)
Hepatocytes/drug effects , Hepatocytes/metabolism , Liver Diseases/genetics , Pyrrolizidine Alkaloids/pharmacology , Animals , Cell Transplantation , Chimera , Disease Models, Animal , Female , Genetic Therapy , Hepatitis B , Hepatitis B virus , Hepatocytes/transplantation , Homeodomain Proteins/genetics , Humans , Hydrolases/genetics , Interleukin Receptor Common gamma Subunit/genetics , Liver/pathology , Liver Diseases/pathology , Malaria , Male , Mice , Mice, Inbred NOD , Mice, Knockout , Plasmodium falciparum
10.
Eur Respir J ; 59(4)2022 04.
Article in English | MEDLINE | ID: mdl-34561286

ABSTRACT

BACKGROUND: Biomarkers of disease progression and treatment response are urgently needed for patients with lymphangioleiomyomatosis (LAM). Activity-based nanosensors, an emerging biosensor class, detect dysregulated proteases in vivo and release a reporter to provide a urinary readout of disease. Because proteases are dysregulated in LAM and may directly contribute to lung function decline, activity-based nanosensors may enable quantitative, real-time monitoring of LAM progression and treatment response. We aimed to assess the diagnostic utility of activity-based nanosensors in a pre-clinical model of pulmonary LAM. METHODS: Tsc2-null cells were injected intravenously into female nude mice to establish a mouse model of pulmonary LAM. A library of 14 activity-based nanosensors, designed to detect proteases across multiple catalytic classes, was administered into the lungs of LAM mice and healthy controls, urine was collected, and mass spectrometry was performed to measure nanosensor cleavage products. Mice were then treated with rapamycin and monitored with activity-based nanosensors. Machine learning was performed to distinguish diseased from healthy and treated from untreated mice. RESULTS: Multiple activity-based nanosensors (PP03 (cleaved by metallo, aspartic and cysteine proteases), padjusted<0.0001; PP10 (cleaved by serine, aspartic and cysteine proteases), padjusted=0.017)) were differentially cleaved in diseased and healthy lungs, enabling strong classification with a machine learning model (area under the curve (AUC) 0.95 from healthy). Within 2 days after rapamycin initiation, we observed normalisation of PP03 and PP10 cleavage, and machine learning enabled accurate classification of treatment response (AUC 0.94 from untreated). CONCLUSIONS: Activity-based nanosensors enable noninvasive, real-time monitoring of disease burden and treatment response in a pre-clinical model of LAM.


Subject(s)
Cysteine Proteases , Lymphangioleiomyomatosis , Animals , Cysteine Proteases/therapeutic use , Female , Humans , Lymphangioleiomyomatosis/drug therapy , Mice , Mice, Nude , Peptide Hydrolases/therapeutic use , Sirolimus/therapeutic use
11.
Nat Mater ; 20(10): 1440-1448, 2021 10.
Article in English | MEDLINE | ID: mdl-34267368

ABSTRACT

Therapeutic outcomes in oncology may be aided by precision diagnostics that offer early detection, localization and the opportunity to monitor response to therapy. Here, we report a multimodal nanosensor engineered to target tumours through acidosis, respond to proteases in the microenvironment to release urinary reporters and (optionally) carry positron emission tomography probes to enable localization of primary and metastatic cancers in mouse models of colorectal cancer. We present a paradigm wherein this multimodal sensor can be employed longitudinally to assess burden of disease non-invasively, including tumour progression and response to chemotherapy. Specifically, we showed that acidosis-mediated tumour insertion enhanced on-target release of matrix metalloproteinase-responsive reporters in urine. Subsequent on-demand loading of the radiotracer 64Cu allowed pH-dependent tumour visualization, enabling enriched microenvironmental characterization when compared with the conventional metabolic tracer 18F-fluorodeoxyglucose. Through tailored target specificities, this modular platform has the capacity to be engineered as a pan-cancer test that may guide treatment decisions for numerous tumour types.


Subject(s)
Acidosis/diagnosis , Colorectal Neoplasms/diagnosis , Multimodal Imaging , Precision Medicine , Tumor Microenvironment , Acidosis/complications , Animals , Colorectal Neoplasms/complications , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Disease Progression , Female , Fluorodeoxyglucose F18 , Mice , Mice, Inbred BALB C , Positron-Emission Tomography
12.
Nature ; 536(7614): 81-85, 2016 08 04.
Article in English | MEDLINE | ID: mdl-27437587

ABSTRACT

The widespread view of bacteria as strictly pathogenic has given way to an appreciation of the prevalence of some beneficial microbes within the human body. It is perhaps inevitable that some bacteria would evolve to preferentially grow in environments that harbor disease and thus provide a natural platform for the development of engineered therapies. Such therapies could benefit from bacteria that are programmed to limit bacterial growth while continually producing and releasing cytotoxic agents in situ. Here we engineer a clinically relevant bacterium to lyse synchronously ata threshold population density and to release genetically encoded cargo. Following quorum lysis, a small number of surviving bacteria reseed the growing population, thus leading to pulsatile delivery cycles. We used microfluidic devices to characterize the engineered lysis strain and we demonstrate its potential as a drug delivery platform via co-culture with human cancer cells in vitro. Asa proof of principle, we tracked the bacterial population dynamics in ectopic syngeneic colorectal tumours in mice via a luminescent reporter. The lysis strain exhibits pulsatile population dynamics in vivo, with mean bacterial luminescence that remained two orders of magnitude lower than an unmodified strain. Finally, guided by previous findings that certain bacteria can enhance the efficacy of standard therapies, we orally administered the lysis strain alone or in combination with a clinical chemotherapeutic to a syngeneic mouse transplantation model of hepatic colorectal metastases. We found that the combination of both circuit-engineered bacteria and chemotherapy leads to a notable reduction of tumour activity along with a marked survival benefit over either therapy alone.Our approach establishes a methodology for leveraging the tools of synthetic biology to exploit the natural propensity for certain bacteria to colonize disease sites.


Subject(s)
Bacteriolysis , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/microbiology , Drug Delivery Systems/methods , Salmonella/metabolism , Administration, Oral , Animals , Coculture Techniques , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Computer Simulation , Female , Liver Neoplasms/secondary , Luminescence , Mice , Neoplasm Metastasis , Neoplasm Transplantation , Quorum Sensing , Salmonella/genetics , Synthetic Biology/methods , Transplantation, Isogeneic
13.
Nature ; 538(7625): 344-349, 2016 Oct 20.
Article in English | MEDLINE | ID: mdl-27602946

ABSTRACT

Antimalarial drugs have thus far been chiefly derived from two sources-natural products and synthetic drug-like compounds. Here we investigate whether antimalarial agents with novel mechanisms of action could be discovered using a diverse collection of synthetic compounds that have three-dimensional features reminiscent of natural products and are underrepresented in typical screening collections. We report the identification of such compounds with both previously reported and undescribed mechanisms of action, including a series of bicyclic azetidines that inhibit a new antimalarial target, phenylalanyl-tRNA synthetase. These molecules are curative in mice at a single, low dose and show activity against all parasite life stages in multiple in vivo efficacy models. Our findings identify bicyclic azetidines with the potential to both cure and prevent transmission of the disease as well as protect at-risk populations with a single oral dose, highlighting the strength of diversity-oriented synthesis in revealing promising therapeutic targets.


Subject(s)
Antimalarials/chemical synthesis , Antimalarials/pharmacology , Azetidines/therapeutic use , Drug Discovery , Life Cycle Stages/drug effects , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Plasmodium falciparum/growth & development , Animals , Antimalarials/administration & dosage , Antimalarials/therapeutic use , Azabicyclo Compounds/administration & dosage , Azabicyclo Compounds/chemical synthesis , Azabicyclo Compounds/pharmacology , Azabicyclo Compounds/therapeutic use , Azetidines/administration & dosage , Azetidines/adverse effects , Azetidines/pharmacology , Cytosol/enzymology , Disease Models, Animal , Female , Liver/drug effects , Liver/parasitology , Macaca mulatta/parasitology , Malaria, Falciparum/prevention & control , Malaria, Falciparum/transmission , Male , Mice , Phenylalanine-tRNA Ligase/antagonists & inhibitors , Phenylurea Compounds/administration & dosage , Phenylurea Compounds/chemical synthesis , Phenylurea Compounds/pharmacology , Phenylurea Compounds/therapeutic use , Plasmodium falciparum/cytology , Plasmodium falciparum/enzymology , Safety
14.
Proc Natl Acad Sci U S A ; 121(11): e2315789121, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38437565
15.
Proc Natl Acad Sci U S A ; 115(36): 8954-8959, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30126988

ABSTRACT

Improved biomarkers are needed for prostate cancer, as the current gold standards have poor predictive value. Tests for circulating prostate-specific antigen (PSA) levels are susceptible to various noncancer comorbidities in the prostate and do not provide prognostic information, whereas physical biopsies are invasive, must be performed repeatedly, and only sample a fraction of the prostate. Injectable biosensors may provide a new paradigm for prostate cancer biomarkers by querying the status of the prostate via a noninvasive readout. Proteases are an important class of enzymes that play a role in every hallmark of cancer; their activities could be leveraged as biomarkers. We identified a panel of prostate cancer proteases through transcriptomic and proteomic analysis. Using this panel, we developed a nanosensor library that measures protease activity in vitro using fluorescence and in vivo using urinary readouts. In xenograft mouse models, we applied this nanosensor library to classify aggressive prostate cancer and to select predictive substrates. Last, we coformulated a subset of nanosensors with integrin-targeting ligands to increase sensitivity. These targeted nanosensors robustly classified prostate cancer aggressiveness and outperformed PSA. This activity-based nanosensor library could be useful throughout clinical management of prostate cancer, with both diagnostic and prognostic utility.


Subject(s)
Biomarkers, Tumor , Gene Expression Profiling , Gene Library , Neoplasms, Experimental , Prostatic Neoplasms , Animals , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Heterografts , Humans , Male , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms, Experimental/classification , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Prostatic Neoplasms/classification , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism
16.
Genes Dev ; 27(22): 2397-408, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24240230

ABSTRACT

Historically, biomedical research has been based on two paradigms. First, measurements of biological behaviors have been based on bulk assays that average over large populations. Second, these behaviors have then been crudely perturbed by systemic administration of therapeutic treatments. Nanotechnology has the potential to transform these paradigms by enabling exquisite structures comparable in size with biomolecules as well as unprecedented chemical and physical functionality at small length scales. Here, we review nanotechnology-based approaches for precisely measuring and perturbing living systems. Remarkably, nanotechnology can be used to characterize single molecules or cells at extraordinarily high throughput and deliver therapeutic payloads to specific locations as well as exhibit dynamic biomimetic behavior. These advances enable multimodal interfaces that may yield unexpected insights into systems biology as well as new therapeutic strategies for personalized medicine.


Subject(s)
Medicine/trends , Nanotechnology , Humans
17.
Gastroenterology ; 156(6): 1788-1804.e13, 2019 05.
Article in English | MEDLINE | ID: mdl-30641053

ABSTRACT

BACKGROUND & AIMS: Patients with cirrhosis are at high risk for hepatocellular carcinoma (HCC) and often have increased serum levels of estrogen. It is not clear how estrogen promotes hepatic growth. We investigated the effects of estrogen on hepatocyte proliferation during zebrafish development, liver regeneration, and carcinogenesis. We also studied human hepatocytes and liver tissues. METHODS: Zebrafish were exposed to selective modifiers of estrogen signaling at larval and adult stages. Liver growth was assessed by gene expression, fluorescent imaging, and histologic analyses. We monitored liver regeneration after hepatocyte ablation and HCC development after administration of chemical carcinogens (dimethylbenzanthrazene). Proliferation of human hepatocytes was measured in a coculture system. We measured levels of G-protein-coupled estrogen receptor (GPER1) in HCC and nontumor liver tissues from 68 patients by immunohistochemistry. RESULTS: Exposure to 17ß-estradiol (E2) increased proliferation of hepatocytes and liver volume and mass in larval and adult zebrafish. Chemical genetic and epistasis experiments showed that GPER1 mediates the effects of E2 via the phosphoinositide 3-kinase-protein kinase B-mechanistic target of rapamycin pathway: gper1-knockout and mtor-knockout zebrafish did not increase liver growth in response to E2. HCC samples from patients had increased levels of GPER1 compared with nontumor tissue samples; estrogen promoted proliferation of human primary hepatocytes. Estrogen accelerated hepatocarcinogenesis specifically in male zebrafish. Chemical inhibition or genetic loss of GPER1 significantly reduced tumor development in the zebrafish. CONCLUSIONS: In an analysis of zebrafish and human liver cells and tissues, we found GPER1 to be a hepatic estrogen sensor that regulates liver growth during development, regeneration, and tumorigenesis. Inhibitors of GPER1 might be developed for liver cancer prevention or treatment. TRANSCRIPT PROFILING: The accession number in the Gene Expression Omnibus is GSE92544.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Estradiol/pharmacology , Estrogens/pharmacology , Liver Neoplasms/metabolism , Liver/growth & development , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism , Zebrafish Proteins/metabolism , 9,10-Dimethyl-1,2-benzanthracene , Animals , Carcinogenesis/drug effects , Carcinoma, Hepatocellular/pathology , Cell Proliferation/drug effects , Female , Gene Expression/drug effects , Hepatocytes , Humans , Liver/metabolism , Liver Cirrhosis/metabolism , Liver Neoplasms/pathology , Liver Regeneration , Male , Organ Size/drug effects , Phosphatidylinositol 3-Kinase/metabolism , Receptors, G-Protein-Coupled/genetics , Sex Factors , Signal Transduction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Tumor Burden/drug effects , Zebrafish , Zebrafish Proteins/genetics
18.
Adv Funct Mater ; 30(48)2020 Nov 25.
Article in English | MEDLINE | ID: mdl-33776613

ABSTRACT

Engineered tissue models comprise a variety of multiplexed ensembles in which combinations of epithelial, stromal, and immune cells give rise to physiologic function. Engineering spatiotemporal control of cell-cell and cell-matrix interactions within these 3D multicellular tissues would represent a significant advance for tissue engineering. In this work, a new method, entitled CAMEO (Controlled Apoptosis in Multicellular tissues for Engineered Organogenesis) enables the non-invasive triggering of controlled apoptosis to eliminate genetically-engineered cells from a pre-established culture. Using this approach, the contribution of stromal cells to the phenotypic stability of primary human hepatocytes is examined. 3D hepatic microtissues, in which fibroblasts can enhance phenotypic stability and accelerate aggregation into spheroids, were found to rely only transiently on fibroblast interaction to support multiple axes of liver function, such as protein secretion and drug detoxification. Due to its modularity, CAMEO has the promise to be readily extendable to other applications that are tied to the complexity of 3D tissue biology, from understanding in vitro organoid models to building artificial tissue grafts.

19.
Adv Funct Mater ; 30(48)2020 Nov 25.
Article in English | MEDLINE | ID: mdl-33613149

ABSTRACT

Formation of capillary blood vasculature is a critical requirement for native as well as engineered organs and can be induced in vitro by co-culturing endothelial cells with fibroblasts. However, whether these fibroblasts are required only in the initial morphogenesis of endothelial cells or needed throughout is unknown, and the ability to remove these stromal cells after assembly could be useful for clinical translation. In this study, we introduce a technique termed CAMEO (Controlled Apoptosis in Multicellular Tissues for Engineered Organogenesis), whereby fibroblasts are selectively ablated on demand, and utilize it to probe the dispensability of fibroblasts in vascular morphogenesis. The presence of fibroblasts is shown to be necessary only during the first few days of endothelial cell morphogenesis, after which they can be ablated without significantly affecting the structural and functional features of the developed vasculature. Furthermore, we demonstrate the use of CAMEO to vascularize a construct containing primary human hepatocytes that improved tissue function. In conclusion, this study suggests that transient, initial support from fibroblasts is sufficient to drive vascular morphogenesis in engineered tissues, and this strategy of engineering-via-elimination may provide a new general approach for achieving desired functions and cell compositions in engineered organs.

20.
Mol Pharm ; 17(9): 3633-3642, 2020 09 08.
Article in English | MEDLINE | ID: mdl-32786959

ABSTRACT

Therapeutic nucleic acids hold great promise for the treatment of genetic diseases, yet the delivery of this highly charged macromolecular drug remains a challenge in the field. Peptides are promising agents to mediate nucleic acid delivery because they can encode a biological function to overcome the trafficking barriers. Electrostatic nanocomplexes of nucleic acid and peptides can achieve effective delivery, but the balance between their stability and biological function must be finely tuned. In this work, we explore two peptide building blocks that have been studied in the literature: targeting ligands and intracellular trafficking peptides. We grafted these peptides on a polyethylene glycol (PEG) backbone with eight sites for substitution to create so-called "peptide spiders". These conjugates achieve stability via the well-known hydrophilic shielding effect of PEG. In addition, the coordination of peptide building blocks into multimers may create new biological properties, such as the well-known phenomena of increased binding avidity with multivalent ligands. In this work, we linked two trafficking peptides to the PEG backbone using either nonreducible or reducible chemistries and investigated the ability of these materials to carry silencing RNAs into mammalian cells. We then investigated these nanomaterials for their pharmacokinetic properties and silencing of undruggable targets in a mouse model of cancer. While reducible linkages were more potent at silencing in vitro, this effect was reversed when applied in the context of living animals. This work offers an insight into peptide-based delivery materials and investigates peptide-polymer linkages.


Subject(s)
Nucleic Acids/chemistry , Peptides/chemistry , Polymers/chemistry , Animals , Cell Line, Tumor , Female , Humans , Ligands , Macromolecular Substances/chemistry , Mice , Mice, Nude , Nanoparticles/chemistry , Neoplasms/metabolism , Polyethylene Glycols/chemistry , RNA, Small Interfering/chemistry , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL